1
|
Hoblos R, Khalil K, Karam M, Bazzi S. The role of NF-κB transcription factor in the regulation of cytokine induced thermal hyperalgesia in a Leishmania major model in BALB/c mice. Exp Parasitol 2024; 267:108864. [PMID: 39577554 DOI: 10.1016/j.exppara.2024.108864] [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: 07/24/2024] [Revised: 11/16/2024] [Accepted: 11/18/2024] [Indexed: 11/24/2024]
Abstract
Cutaneous leishmaniasis caused mainly by Leishmania major (L. major) is one of the trending models used to investigate induced hyperalgesia and the involved cytokines. Previous studies approached the role of several cytokines in the observed hyperalgesia, but the molecular mechanisms orchestrating such a response still needed to be addressed. In this study, we inspect the role of the NF-κB in the modulation of L. major-prompted hyperalgesia and cytokine expression in BALB/c mice by administering celastrol, a potent blocker of this transcription factor. Intraperitoneal injection of 0.5 mg/kg and 1 mg/kg of celastrol attenuated the L. major-induced thermal hyperalgesia in BALB/c mice for 15 days and 21 days, respectively, as detected by hot plate and tail flick behavioral assessments. Cytokine levels were quantified in the infected paws of BALB/c mice using Sandwich ELISA. The administration of 1 mg/kg celastrol decreased TNF-α levels in L. major infected mice for 23 days, and IL-1β expression declined significantly for 23 days using both celastrol dosages. However, no significant change was observed in the levels of IL-10 in our experimental groups. The activation of NF-κB was detected by observing the phosphorylation levels of the p65 subunit using PathScan phospho-ELISA. The level of NF-κB phosphorylation was elevated in L. major infected BALB/c mice. Only administering 1 mg/kg celastrol suppressed the phosphorylation of p65, thus inactivating NF-kB. In conclusion, our results provide new insights into the correlation between the activation of NF-kB, the induction of thermal hyperalgesia, and the expression of TNF-α and IL-1β in the L. major-induced hyperalgesia model.
Collapse
Affiliation(s)
- Reem Hoblos
- University of Balamand, Faculty of Arts and Sciences, Lebanon
| | - Karl Khalil
- Lebanese American University, School of Medicine, Lebanon.
| | - Marc Karam
- University of Balamand, Faculty of Arts and Sciences, Lebanon
| | - Samer Bazzi
- University of Balamand, Faculty of Arts and Sciences, Lebanon
| |
Collapse
|
2
|
Uygun H, Oren AC, Sahinoglu EP, Akbayram S. Recurrent Visceral Leishmaniasis in a Case With Interleukin-12 Receptor Beta-1 Deficiency. Acta Parasitol 2024; 69:2069-2072. [PMID: 39388053 DOI: 10.1007/s11686-024-00926-8] [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: 03/29/2024] [Accepted: 09/11/2024] [Indexed: 10/12/2024]
Abstract
PURPOSE In this study, we present the case of a children who was followed up for recurrent visceral leishmaniasis and diagnosed with IL-12Rβ1 deficiency. METHODS A female patient who received Bacille Calmette-Guérin (BCG) vaccine 2 months after birth and developed visceral leishmaniasis at the age of 91 months was subsequently diagnosed with IL-12Rβ1 deficiency. The patient's diagnosis and treatment process were examined retrospectively. RESULTS IL-12Rβ1 deficiency is an autosomal recessive disease characterized by susceptibility to recurrent and/or severe infections caused by weakly pathogenic mycobacteria and salmonella. Infections with other intramacrophagic organisms may also occur, although rarely. Based on this information, it is believed that the mutation in the IFN-γ/IL-12 axis in our patient predisposed her to recurrent Leishmania infections. CONCLUSION This study adds to the limited literature on IL12RB1 deficiency as a cause of VL. Patients diagnosed with VL should be evaluated immunologically, as recurrent Leishmania infections may occur in those with IL-12Rβ1 defects.
Collapse
Affiliation(s)
- Hatice Uygun
- Department of Pediatric Infectious Disease, Gaziantep University School of Medicine, Gaziantep, Turkey.
| | - Ayse Ceyda Oren
- Department of Pediatric Hematology and Oncology, Gaziantep Liv Hospital, Gaziantep, Turkey
| | - Esra Pekpak Sahinoglu
- Department of Pediatric Hematology and Oncology, Gaziantep Liv Hospital, Gaziantep, Turkey
| | - Sinan Akbayram
- Department of Pediatric Hematology and Oncology, Gaziantep Liv Hospital, Gaziantep, Turkey
| |
Collapse
|
3
|
Sanz CR, Sarquis J, Daza MÁ, Miró G. Exploring the impact of epidemiological and clinical factors on the progression of canine leishmaniosis by statistical and whole genome analyses: from breed predisposition to comorbidities. Int J Parasitol 2024; 54:401-414. [PMID: 38570155 DOI: 10.1016/j.ijpara.2024.03.006] [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: 07/14/2023] [Revised: 02/25/2024] [Accepted: 03/26/2024] [Indexed: 04/05/2024]
Abstract
Canine leishmaniosis (CanL), caused by Leishmania infantum, is a complex disease of growing importance in Europe. Clinical manifestations result from the down-modulation of the host immune response through multiple host-parasite interactions. Although several factors might influence CanL progression, this is the first known study evaluating risk factors for its different clinical stages in a large referral hospital population (n = 35.669) from an endemic area, over a 20 year period. Genome-wide scans for selection signatures were also conducted to explore the genomic component of clinical susceptibility to L. infantum infection. The prevalence of CanL was 3.2% (16.7% stage I; 43.6% stage II; 32.1% stage III; 7.6% stage IV). Dog breed (crossbreed), bodyweight (<10 kg), living conditions (indoors), regular deworming treatment, and being vaccinated against Leishmania significantly decreased the transmission risk and the risk for developing severe clinical forms. Conversely, the detection of comorbidities was associated with advanced clinical forms, particularly chronic kidney disease, neoplasia, cryptorchidism, infectious tracheobronchitis and urate urolithiasis, although those did not impact the clinical outcome. Significant associations between an increased risk of severe clinical stages and findings in the anamnesis (renal or skin-related manifestations) and physical examination (ocular findings) were also detected, highlighting their diagnostic value in referred cases of CanL. Sixteen breeds were found to be significantly more susceptible to developing severe stages of leishmaniosis (e.g. Great Dane, Rottweiler, English Springer Spaniel, Boxer, American Staffordshire Terrier, Golden Retriever), while 20 breeds displayed a clinical resistantance phenotype and, thus, are more likely to mount an efficient immune response against L. infantum (e.g. Pointer, Samoyed, Spanish Mastiff, Spanish Greyhound, English Setter, Siberian Husky). Genomic analyses of these breeds retrieved 12 regions under selection, 63 candidate genes and pinpointed multiple biological pathways such as the IRE1 branch of the unfolded protein response, which could play a critical role in clinical susceptibility to L. infantum infection.
Collapse
Affiliation(s)
- Carolina R Sanz
- Animal Health Department, Veterinary Faculty, Complutense University of Madrid, Av. Puerta de Hierro s/n, Madrid 28040, Spain.
| | - Juliana Sarquis
- Animal Health Department, Veterinary Faculty, Complutense University of Madrid, Av. Puerta de Hierro s/n, Madrid 28040, Spain
| | - María Ángeles Daza
- Veterinary Teaching Hospital, Veterinary Faculty, Complutense University of Madrid, Av. Puerta Hierro s/n, Madrid 28040, Spain; Department of Animal Medicine and Surgery, Veterinary Faculty, Complutense University of Madrid, Av. Puerta de Hierro s/n, Madrid 28040, Spain
| | - Guadalupe Miró
- Animal Health Department, Veterinary Faculty, Complutense University of Madrid, Av. Puerta de Hierro s/n, Madrid 28040, Spain; Veterinary Teaching Hospital, Veterinary Faculty, Complutense University of Madrid, Av. Puerta Hierro s/n, Madrid 28040, Spain.
| |
Collapse
|
4
|
Dirkx L, Van Acker SI, Nicolaes Y, Cunha JLR, Ahmad R, Hendrickx R, Caljon B, Imamura H, Ebo DG, Jeffares DC, Sterckx YGJ, Maes L, Hendrickx S, Caljon G. Long-term hematopoietic stem cells trigger quiescence in Leishmania parasites. PLoS Pathog 2024; 20:e1012181. [PMID: 38656959 PMCID: PMC11073788 DOI: 10.1371/journal.ppat.1012181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 05/06/2024] [Accepted: 04/09/2024] [Indexed: 04/26/2024] Open
Abstract
Addressing the challenges of quiescence and post-treatment relapse is of utmost importance in the microbiology field. This study shows that Leishmania infantum and L. donovani parasites rapidly enter into quiescence after an estimated 2-3 divisions in both human and mouse bone marrow stem cells. Interestingly, this behavior is not observed in macrophages, which are the primary host cells of the Leishmania parasite. Transcriptional comparison of the quiescent and non-quiescent metabolic states confirmed the overall decrease of gene expression as a hallmark of quiescence. Quiescent amastigotes display a reduced size and signs of a rapid evolutionary adaptation response with genetic alterations. Our study provides further evidence that this quiescent state significantly enhances resistance to treatment. Moreover, transitioning through quiescence is highly compatible with sand fly transmission and increases the potential of parasites to infect cells. Collectively, this work identified stem cells in the bone marrow as a niche where Leishmania quiescence occurs, with important implications for antiparasitic treatment and acquisition of virulence traits.
Collapse
Affiliation(s)
- Laura Dirkx
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Sara I. Van Acker
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Yasmine Nicolaes
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - João Luís Reis Cunha
- York Biomedical Research Institute and Department of Biology, University of York, York, United Kingdom
| | - Rokaya Ahmad
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Rik Hendrickx
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Ben Caljon
- Brussels Interuniversity Genomics High Throughput core (BRIGHTcore) platform, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Hideo Imamura
- Brussels Interuniversity Genomics High Throughput core (BRIGHTcore) platform, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Didier G. Ebo
- Department of Immunology–Allergology–Rheumatology, Faculty of Medicine and Health Science, Infla-Med Centre of Excellence, University of Antwerp, Antwerp University Hospital, Antwerp, Belgium
| | - Daniel C. Jeffares
- York Biomedical Research Institute and Department of Biology, University of York, York, United Kingdom
| | - Yann G.-J. Sterckx
- Laboratory of Medical Biochemistry (LMB), Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Louis Maes
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Sarah Hendrickx
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Guy Caljon
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
5
|
Yin S, Li J, Chen J, Zhou Q, Duan DBP, Lai M, Zhong J, He J, Chen D, Zeng Z, Su L, Luo L, Dong C, Zheng Z. LdCyPA attenuates MAPK pathway to assist Leishmania donovani immune escape in host cells. Acta Trop 2024; 251:107114. [PMID: 38190929 DOI: 10.1016/j.actatropica.2023.107114] [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: 11/07/2023] [Revised: 12/20/2023] [Accepted: 12/26/2023] [Indexed: 01/10/2024]
Abstract
BACKGROUND Visceral leishmaniasis is a neglected tropical disease affecting millions of people worldwide. Macrophages serve as the primary host cells for L. donovani, the immune response capability of these host cells is crucial for parasites' intracellular survival. L. donovani peptidyl-prolyl cis/trans isomerase Cyclophilin A (LdCypA) is a key protein for L. donovani intracellular proliferation, while the molecular mechanism conducive to intracellular survival of parasites remains elusive. METHODS In this study, we generated a macrophage cell line overexpressing LdCyPA to investigate its role in controlling host immunity and promoting intracellular immune escape of L. donovani. RESULTS It was discovered that the overexpression of the LdCyPA cell line regulated the host immune response following infection by downregulating the proportion of M1-type macrophages, promoting the secretion of the anti-inflammatory factor IL-4, and inhibiting the secretion of pro-inflammatory factors like IL-12, IFN-γ, TNF-α, and INOS. Transcriptome sequencing and mechanistic validation, meanwhile, demonstrated that cells overexpressing LdCyPA controlled the immune responses that followed infection by blocking the phosphorylation of P38 and JNK1/2 proteins in the MAPK signaling pathway and simultaneously increasing the phosphorylation of ERK proteins, which helped the L. donovani escape immune recognition. CONCLUSION Our findings thus pave the way for the development of host-directed antiparasitic drugs by illuminating the pro-Leishmania survival mechanism of L. donovani cyclophilin A and exposing a novel immune escape strategy for L. donovani that targets host cellular immune regulation.
Collapse
Affiliation(s)
- Shuangshuang Yin
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, PR China
| | - Jiao Li
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, PR China; Sichuan-Chongqing jointly-established Research Platform of Zoonosis, Chengdu, PR China
| | - Jianping Chen
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, PR China; Sichuan-Chongqing jointly-established Research Platform of Zoonosis, Chengdu, PR China
| | - Qi Zhou
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, PR China
| | - Deng Bin Pei Duan
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, PR China
| | - Meng Lai
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, PR China
| | - Junchao Zhong
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, PR China
| | - Jinlei He
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, PR China; Sichuan-Chongqing jointly-established Research Platform of Zoonosis, Chengdu, PR China
| | - Dali Chen
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, PR China; Sichuan-Chongqing jointly-established Research Platform of Zoonosis, Chengdu, PR China
| | - Zheng Zeng
- Sichuan-Chongqing jointly-established Research Platform of Zoonosis, Chengdu, PR China; Chong Qing Animal Disease Prevention and Control Center, Chongqing, PR China
| | - Liang Su
- Sichuan-Chongqing jointly-established Research Platform of Zoonosis, Chengdu, PR China; Chong Qing Animal Disease Prevention and Control Center, Chongqing, PR China
| | - Lu Luo
- Sichuan-Chongqing jointly-established Research Platform of Zoonosis, Chengdu, PR China; Chong Qing Animal Disease Prevention and Control Center, Chongqing, PR China
| | - Chunxia Dong
- Sichuan-Chongqing jointly-established Research Platform of Zoonosis, Chengdu, PR China; Chong Qing Animal Disease Prevention and Control Center, Chongqing, PR China
| | - Zhiwan Zheng
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, PR China; Sichuan-Chongqing jointly-established Research Platform of Zoonosis, Chengdu, PR China.
| |
Collapse
|
6
|
Mulè A, Crosato V, Kuhns DB, Lorenzi L, Chirico C, Maifredi G, Notarangelo LD, Castelli F, Tomasoni LR. Visceral Leishmaniasis in Immunocompetent Hosts in Brescia: A Case Series and Analysis of Cytokine Cascade. Microorganisms 2024; 12:394. [PMID: 38399799 PMCID: PMC10892745 DOI: 10.3390/microorganisms12020394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 02/06/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Visceral leishmaniasis (VL) is a parasitic zoonosis caused by Leishmania spp. that usually manifests itself in immunocompromised subjects. It is a rare and neglected disease, and it is not endemic in the province of Brescia (Italy). Three cases of human VL occurred in Brescia from October to December 2021 in immunocompetent patients. We evaluated the patients looking for signs of underlying immunodeficiencies and conducted further epidemiological evaluations in the province of Brescia without success. An analysis of the sera levels of the main cytokines involved in the immune response to VL was performed. All patients presented a significant augmentation of CXCL-10, CCL-4, and IL-6. The patients tested during the acute phase showed an elevation of IL-1α, IL-5, IL-10, and IL-12, while in the recovery phase, higher levels of TNF-α and IL-7 were detected. Altogether, a predominant activation of the T-helper-2 pathway emerged during the acute phase of the parasite infection, while the cytokines associated with the T-helper-1 pathway were less represented. This imbalanced immune response to the parasite infection might play a crucial role in the development of VL in immunocompetent patients.
Collapse
Affiliation(s)
- Alice Mulè
- Unit of Infectious and Tropical Diseases, Department of Clinical and Experimental Sciences, University of Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy; (V.C.); (F.C.)
| | - Verena Crosato
- Unit of Infectious and Tropical Diseases, Department of Clinical and Experimental Sciences, University of Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy; (V.C.); (F.C.)
| | - Douglas Byron Kuhns
- Neutrophil Monitoring Laboratory, Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD 20701, USA;
| | - Luisa Lorenzi
- Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy;
| | - Claudia Chirico
- Department of Hygiene and Health Prevention, Health Protection Agency of Brescia (ATS Brescia), 25124 Brescia, Italy
| | - Giovanni Maifredi
- Epidemiology Unit, Health Protection Agency of Brescia (ATS Brescia), 25124 Brescia, Italy;
| | - Luigi D. Notarangelo
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, NIAID, NIH, Bethesda, MD 20852, USA;
| | - Francesco Castelli
- Unit of Infectious and Tropical Diseases, Department of Clinical and Experimental Sciences, University of Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy; (V.C.); (F.C.)
| | - Lina R. Tomasoni
- Unit of Infectious and Tropical Diseases, ASST Spedali Civili di Brescia, 25123 Brescia, Italy;
| |
Collapse
|
7
|
Saini S, Anand A, Singh A, Mahapatra B, Sirohi S, Singh S, Singh RK. Swarna Bhasma Induces Antigen-Presenting Abilities of Macrophages and Helps Antigen Experienced CD4 + T Cells to Acquire Th1 Phenotypes Against Leishmania donovani Antigens. Biol Trace Elem Res 2024; 202:210-220. [PMID: 37088826 PMCID: PMC10123016 DOI: 10.1007/s12011-023-03659-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 04/03/2023] [Indexed: 04/25/2023]
Abstract
In leishmaniasis, the protective immunity is largely mediated by proinflammatory cytokine producing abilities of T cells and an efficient parasite killing by phagocytic cells. Notwithstanding a substantial progress that has been made during last decades, the mechanisms or factors involved in establishing protective immunity against Leishmania are not identified. In ancient Indian literature, metallic "bhasma," particularly that of "swarna" or gold (fine gold particles), is indicated as one of the most prominent metal-based therapeutic medicine, which is known to impart protective and curative properties in various health issues. In this work, we elucidated the potential of swarna bhasma (SB) on the effector properties of phagocytes and antigen-activated CD4+ T cells in augmenting the immunogenicity of L. donovani antigens. The characterization of SB revealing its shape, size, composition, and measurement of cytotoxicity established the physiochemical potential for its utilization as an immunomodulator. The activation of macrophages with SB enhanced their capacity to produce nitric oxide and proinflammatory cytokines, which eventually resulted in reduced uptake of parasites and their proliferation in infected cells. Further, in Leishmania-infected animals, SB administration reduced the generation of IL-10, an anti-inflammatory cytokine, and enhanced pro-inflammatory cytokine generation by antigen activated CD4+ T cells with increased frequency of double (IFNγ+/TNFα+) and triple (IFNγ+TNFα+IL-2+) positive cells and abrogated disease pathogeneses at the early days of infection. Our results also suggested that cow-ghee (A2) emulsified preparation of SB, either alone or with yashtimadhu, a known natural immune modulator which enhances the SB's potential in enhancing the immunogenicity of parasitic antigens. These findings suggested a definite potential of SB in enhancing the effector functions of phagocytes and CD4+ T cells against L. donovani antigens. Therefore, more studies are needed to elucidate the mechanistic details of SB and its potential in enhancing vaccine-induced immunity.
Collapse
Affiliation(s)
- Shashi Saini
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221 005, India
| | - Anshul Anand
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221 005, India
| | - Abhishek Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221 005, India
| | - Baishakhi Mahapatra
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221 005, India
| | - Shruti Sirohi
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221 005, India
| | - Samer Singh
- Centre of Experimental Medicine and Surgery, Institute of Medical Science, Banaras Hindu University, Varanasi, 221 005, India
| | - Rakesh K Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221 005, India.
| |
Collapse
|
8
|
Veras PST, de Santana MBR, Brodskyn CI, Fraga DBM, Solcà MS, De Menezes JPB, Leite BMM, Teixeira HMP. Elucidating the role played by bone marrow in visceral leishmaniasis. Front Cell Infect Microbiol 2023; 13:1261074. [PMID: 37860064 PMCID: PMC10582953 DOI: 10.3389/fcimb.2023.1261074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/08/2023] [Indexed: 10/21/2023] Open
Abstract
Leishmaniasis is a widespread group of infectious diseases that significantly impact global health. Despite high prevalence, leishmaniasis often receives inadequate attention in the prioritization of measures targeting tropical diseases. The causative agents of leishmaniasis are protozoan parasites of the Leishmania genus, which give rise to a diverse range of clinical manifestations, including cutaneous and visceral forms. Visceral leishmaniasis (VL), the most severe form, can be life-threatening if left untreated. Parasites can spread systemically within the body, infecting a range of organs, such as the liver, spleen, bone marrow and lymph nodes. Natural reservoirs for these protozoa include rodents, dogs, foxes, jackals, and wolves, with dogs serving as the primary urban reservoir for Leishmania infantum. Dogs exhibit clinical and pathological similarities to human VL and are valuable models for studying disease progression. Both human and canine VL provoke clinical symptoms, such as organ enlargement, fever, weight loss and abnormal gamma globulin levels. Hematologic abnormalities have also been observed, including anemia, leukopenia with lymphocytosis, neutropenia, and thrombocytopenia. Studies in dogs have linked these hematologic changes in peripheral blood to alterations in the bone marrow. Mouse models of VL have also contributed significantly to our understanding of the mechanisms underlying these hematologic and bone marrow abnormalities. This review consolidates information on hematological and immunological changes in the bone marrow of humans, dogs, and mice infected with Leishmania species causing VL. It includes findings on the role of bone marrow as a source of parasite persistence in internal organs and VL development. Highlighting gaps in current knowledge, the review emphasizes the need for future research to enhance our understanding of VL and identify potential targets for novel diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Patricia Sampaio Tavares Veras
- Laboratory of Parasite - Host Interaction and Epidemiology, Gonçalo Moniz Institute-Fiocruz Bahia, Salvador, Bahia, Brazil
- National Institute of Science and Technology of Tropical Diseases, National Council for Scientific Research and Development (CNPq), Salvador, Brazil
| | - Maria Borges Rabêlo de Santana
- Laboratory of Parasite - Host Interaction and Epidemiology, Gonçalo Moniz Institute-Fiocruz Bahia, Salvador, Bahia, Brazil
| | - Claudia Ida Brodskyn
- Laboratory of Parasite - Host Interaction and Epidemiology, Gonçalo Moniz Institute-Fiocruz Bahia, Salvador, Bahia, Brazil
| | - Deborah Bittencourt Mothé Fraga
- Laboratory of Parasite - Host Interaction and Epidemiology, Gonçalo Moniz Institute-Fiocruz Bahia, Salvador, Bahia, Brazil
- Department of Preventive Veterinary Medicine and Animal Production, School of Veterinary Medicine and Animal Science, Federal University of Bahia, Salvador, Brazil
| | - Manuela Silva Solcà
- Laboratory of Parasite - Host Interaction and Epidemiology, Gonçalo Moniz Institute-Fiocruz Bahia, Salvador, Bahia, Brazil
- Department of Preventive Veterinary Medicine and Animal Production, School of Veterinary Medicine and Animal Science, Federal University of Bahia, Salvador, Brazil
| | | | - Bruna Martins Macedo Leite
- Laboratory of Parasite - Host Interaction and Epidemiology, Gonçalo Moniz Institute-Fiocruz Bahia, Salvador, Bahia, Brazil
| | | |
Collapse
|
9
|
Volpedo G, Pacheco-Fernandez T, Oljuskin T, Markle HL, Azodi N, Hamano S, Matlashewski G, Gannavaram S, Nakhasi HL, Satoskar AR. Leishmania mexicana centrin knockout parasites promote M1-polarizing metabolic changes. iScience 2023; 26:107594. [PMID: 37744404 PMCID: PMC10517399 DOI: 10.1016/j.isci.2023.107594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 06/07/2023] [Accepted: 08/07/2023] [Indexed: 09/26/2023] Open
Abstract
Leishmaniasis is a tropical disease prevalent in 90 countries. Presently, there is no approved vaccine for human use. We developed a live attenuated L. mexicana Cen-/-(LmexCen-/-) strain as a vaccine candidate that showed excellent efficacy, characterized by reduced Th2 and enhanced Th1 responses in C57BL/6 and BALB/c mice, respectively, compared to wild-type L. mexicana (LmexWT) infection. Toward understanding the immune mechanisms of protection, we applied untargeted mass spectrometric analysis to LmexCen-/- and LmexWT infections. Data showed enrichment of the pentose phosphate pathway (PPP) in ears immunized with LmexCen-/-versus naive and LmexWT infection. PPP promotes M1 polarization in macrophages, suggesting a switch to a pro-inflammatory phenotype following LmexCen-/- inoculation. Accordingly, PPP inhibition in macrophages infected with LmexCen-/- reduced the production of nitric oxide and interleukin (IL)-1β, hallmarks of classical activation. Overall, our study revealed the immune regulatory mechanisms that may be critical for the induction of protective immunity.
Collapse
Affiliation(s)
- Greta Volpedo
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Thalia Pacheco-Fernandez
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
- Division of Emerging and Transfusion Transmitted Diseases, CBER, FDA, Silver Spring, MD, USA
| | - Timur Oljuskin
- Division of Emerging and Transfusion Transmitted Diseases, CBER, FDA, Silver Spring, MD, USA
| | - Hannah L. Markle
- Division of Emerging and Transfusion Transmitted Diseases, CBER, FDA, Silver Spring, MD, USA
| | - Nazli Azodi
- Division of Emerging and Transfusion Transmitted Diseases, CBER, FDA, Silver Spring, MD, USA
| | - Shinjiro Hamano
- Department of Parasitology, Institute of Tropical Medicine (NEKKEN), The Joint Usage/Research Center on Tropical Disease, Nagasaki University, Nagasaki, Japan
- Nagasaki University Graduate School of Biomedical Sciences Doctoral Leadership Program, Nagasaki, Japan
| | - Greg Matlashewski
- Department of Microbiology and Immunology, McGill University, Montreal, Canada
| | - Sreenivas Gannavaram
- Division of Emerging and Transfusion Transmitted Diseases, CBER, FDA, Silver Spring, MD, USA
| | - Hira L. Nakhasi
- Division of Emerging and Transfusion Transmitted Diseases, CBER, FDA, Silver Spring, MD, USA
| | - Abhay R. Satoskar
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
10
|
Khandibharad S, Singh S. Immuno-metabolic signaling in leishmaniasis: insights gained from mathematical modeling. BIOINFORMATICS ADVANCES 2023; 3:vbad125. [PMID: 37799190 PMCID: PMC10548086 DOI: 10.1093/bioadv/vbad125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/27/2023] [Accepted: 09/12/2023] [Indexed: 10/07/2023]
Abstract
Motivation Leishmaniasis is a global concern especially in underdeveloped and developing subtropical and tropical regions. The extent of infectivity in host is majorly dependent on functional polarization of macrophages. Classically activated M1 macrophage can eliminate parasite through production of iNOS and alternatively activated M2 macrophages can promote parasite growth through by providing shelter and nutrients to parasite. The biological processes involved in immune signaling and metabolism of host and parasite might be responsible for deciding fate of parasite. Results Using systems biology approach, we constructed two mathematical models and inter-regulatory immune-metabolic networks of M1 and M2 state, through which we identified crucial components that are associated with these phenotypes. We also demonstrated how parasite may modulate M1 phenotype for its growth and proliferation and transition to M2 state. Through our previous findings as well as from recent findings we could identify SHP-1 as a key component in regulating the immune-metabolic characterization of M2 macrophage. By targeting SHP-1 at cellular level, it might be possible to modulate immuno-metabolic mechanism and thereby control parasite survival. Availability and implementation Mathematical modeling is implemented as a workflow and the models are deposited in BioModel database. FactoMineR is available at: https://github.com/cran/FactoMineR/tree/master.
Collapse
Affiliation(s)
- Shweta Khandibharad
- Systems Medicine Laboratory, National Centre for Cell Science, NCCS Complex, SPPU Campus, Pune 411007, India
| | - Shailza Singh
- Systems Medicine Laboratory, National Centre for Cell Science, NCCS Complex, SPPU Campus, Pune 411007, India
| |
Collapse
|
11
|
Roy S, Gupta AK, Banerjee M, Das PK, Ukil A. PD-1 negatively tunes macrophage immune activation by turning off JNK and STAT1 signaling: Exploited by Leishmania for its intra-macrophage survival. Cell Immunol 2023; 391-392:104758. [PMID: 37651886 DOI: 10.1016/j.cellimm.2023.104758] [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: 06/16/2023] [Revised: 08/02/2023] [Accepted: 08/21/2023] [Indexed: 09/02/2023]
Abstract
The anti-inflammatory role of the programmed death-1 receptor (PD-1) is well appreciated. However, the mechanism of how PD-1 signaling inhibits the pro-inflammatory cytokine responses in macrophages, which is further exploited by Leishmania to foster their intracellular survival, was unknown. We found that among three major MAP kinases regulating immune activation, PD-1 signaling decreased only JNK phosphorylation without perturbing p38 and ERK. Inflammatory transcription factor STAT1 was also inhibited by PD-1. Association studies documented that SHP, the downstream phosphatase of PD-1, is directly responsible for the decreased phosphorylation of JNK and STAT1. JNK and STAT1 deactivation led to Elk-1/c-Fos inhibition, which significantly decreased IL-12 and TNF-α levels. Further investigation revealed c-Fos deactivation ultimately rendered transcription factor AP1 inactive and facilitating parasite-favorable anti-inflammatory environment.
Collapse
Affiliation(s)
- Shalini Roy
- CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Kolkata 700032, India
| | - Anand K Gupta
- CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Kolkata 700032, India
| | - Madhurima Banerjee
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, India
| | - Pijush K Das
- CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Kolkata 700032, India.
| | - Anindita Ukil
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, India.
| |
Collapse
|
12
|
Valigurová A, Kolářová I. Unrevealing the Mystery of Latent Leishmaniasis: What Cells Can Host Leishmania? Pathogens 2023; 12:pathogens12020246. [PMID: 36839518 PMCID: PMC9967396 DOI: 10.3390/pathogens12020246] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023] Open
Abstract
Leishmania spp. (Kinetoplastida) are unicellular parasites causing leishmaniases, neglected tropical diseases of medical and veterinary importance. In the vertebrate host, Leishmania parasites multiply intracellularly in professional phagocytes, such as monocytes and macrophages. However, their close relative with intracellular development-Trypanosoma cruzi-can unlock even non-professional phagocytes. Since Leishmania and T. cruzi have similar organelle equipment, is it possible that Leishmania can invade and even proliferate in cells other than the professional phagocytes? Additionally, could these cells play a role in the long-term persistence of Leishmania in the host, even in cured individuals? In this review, we provide (i) an overview of non-canonical Leishmania host cells and (ii) an insight into the strategies that Leishmania may use to enter them. Many studies point to fibroblasts as already established host cells that are important in latent leishmaniasis and disease epidemiology, as they support Leishmania transformation into amastigotes and even their multiplication. To invade them, Leishmania causes damage to their plasma membrane and exploits the subsequent repair mechanism via lysosome-triggered endocytosis. Unrevealing the interactions between Leishmania and its non-canonical host cells may shed light on the persistence of these parasites in vertebrate hosts, a way to control latent leishmaniasis.
Collapse
Affiliation(s)
- Andrea Valigurová
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, 611 37 Brno, Czech Republic
- Correspondence: (A.V.); (I.K.)
| | - Iva Kolářová
- Department of Parasitology, Faculty of Science, Charles University, Albertov 6, 128 44 Prague, Czech Republic
- Correspondence: (A.V.); (I.K.)
| |
Collapse
|
13
|
Bandi C, Mendoza-Roldan JA, Otranto D, Alvaro A, Louzada-Flores VN, Pajoro M, Varotto-Boccazzi I, Brilli M, Manenti A, Montomoli E, Zuccotti G, Epis S. Leishmania tarentolae: a vaccine platform to target dendritic cells and a surrogate pathogen for next generation vaccine research in leishmaniases and viral infections. Parasit Vectors 2023; 16:35. [PMID: 36703216 PMCID: PMC9879565 DOI: 10.1186/s13071-023-05651-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 01/03/2023] [Indexed: 01/27/2023] Open
Abstract
Parasites of the genus Leishmania are unusual unicellular microorganisms in that they are characterized by the capability to subvert in their favor the immune response of mammalian phagocytes, including dendritic cells. Thus, in overt leishmaniasis, dendritic cells and macrophages are converted into a niche for Leishmania spp. in which the parasite, rather than being inactivated and disassembled, survives and replicates. In addition, Leishmania parasites hitchhike onto phagocytic cells, exploiting them as a mode of transport to lymphoid tissues where other phagocytic cells are potentially amenable to parasite colonization. This propensity of Leishmania spp. to target dendritic cells has led some researchers to consider the possibility that the non-pathogenic, reptile-associated Leishmania tarentolae could be exploited as a vaccine platform and vehicle for the production of antigens from different viruses and for the delivery of the antigens to dendritic cells and lymph nodes. In addition, as L. tarentolae can also be regarded as a surrogate of pathogenic Leishmania parasites, this parasite of reptiles could possibly be developed into a vaccine against human and canine leishmaniases, exploiting its immunological cross-reactivity with other Leishmania species, or, after its engineering, for the expression of antigens from pathogenic species. In this article we review published studies on the use of L. tarentolae as a vaccine platform and vehicle, mainly in the areas of leishmaniases and viral infections. In addition, a short summary of available knowledge on the biology of L. tarentolae is presented, together with information on the use of this microorganism as a micro-factory to produce antigens suitable for the serodiagnosis of viral and parasitic infections.
Collapse
Affiliation(s)
- Claudio Bandi
- grid.4708.b0000 0004 1757 2822Department of Biosciences, Pediatric CRC “Romeo ed Enrica Invernizzi”–University of Milan, Milan, Italy
| | | | - Domenico Otranto
- grid.7644.10000 0001 0120 3326Department of Veterinary Medicine, University of Bari, Valenzano, Italy
| | - Alessandro Alvaro
- grid.4708.b0000 0004 1757 2822Department of Biosciences, Pediatric CRC “Romeo ed Enrica Invernizzi”–University of Milan, Milan, Italy
| | | | - Massimo Pajoro
- grid.4708.b0000 0004 1757 2822Department of Biosciences, Pediatric CRC “Romeo ed Enrica Invernizzi”–University of Milan, Milan, Italy
| | - Ilaria Varotto-Boccazzi
- grid.4708.b0000 0004 1757 2822Department of Biosciences, Pediatric CRC “Romeo ed Enrica Invernizzi”–University of Milan, Milan, Italy
| | - Matteo Brilli
- grid.4708.b0000 0004 1757 2822Department of Biosciences, Pediatric CRC “Romeo ed Enrica Invernizzi”–University of Milan, Milan, Italy
| | | | - Emanuele Montomoli
- grid.511037.1VisMederi, Siena, Italy ,grid.9024.f0000 0004 1757 4641Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Gianvincenzo Zuccotti
- grid.4708.b0000 0004 1757 2822Department of Biomedical and Clinical Sciences, Pediatric CRC “Romeo ed Enrica Invernizzi”–University of Milan, Milan, Italy ,Department of Pediatrics, Ospedale dei Bambini-Buzzi, Milan, Italy
| | - Sara Epis
- grid.4708.b0000 0004 1757 2822Department of Biosciences, Pediatric CRC “Romeo ed Enrica Invernizzi”–University of Milan, Milan, Italy
| |
Collapse
|
14
|
Dhanalakshmi M, Sruthi D, Jinuraj KR, Das K, Dave S, Andal NM, Das J. Mannose: a potential saccharide candidate in disease management. Med Chem Res 2023; 32:391-408. [PMID: 36694836 PMCID: PMC9852811 DOI: 10.1007/s00044-023-03015-z] [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: 11/05/2022] [Accepted: 01/04/2023] [Indexed: 01/21/2023]
Abstract
There are a plethora of antibiotic resistance cases and humans are marching towards another big survival test of evolution along with drastic climate change and infectious diseases. Ever since the first antibiotic [penicillin], and the myriad of vaccines, we were privileged to escape many infectious disease threats. The survival technique of pathogens seems rapidly changing and sometimes mimicking our own systems in such a perfect manner that we are left unarmed against them. Apart from searching for natural alternatives, repurposing existing drugs more effectively is becoming a familiar approach to new therapeutic opportunities. The ingenious use of revolutionary artificial intelligence-enabled drug discovery techniques is coping with the speed of such alterations. D-Mannose is a great hope as a nutraceutical in drug discovery, against CDG, diabetes, obesity, lung disease, and autoimmune diseases and recent findings of anti-tumor activity make it interesting along with its role in drug delivery enhancing techniques. A very unique work done in the present investigation is the collection of data from the ChEMBL database and presenting the targetable proteins on pathogens as well as on humans. It shows Mannose has 50 targets and the majority of them are on human beings. The structure and conformation of certain monosaccharides have a decisive role in receptor pathogen interactions and here we attempt to review the multifaceted roles of Mannose sugar, its targets associated with different diseases, as a natural molecule having many success stories as a drug and future hope for disease management. Graphical abstract
Collapse
Affiliation(s)
- M. Dhanalakshmi
- Research and Development Centre, Bharathiar University, Coimbatore, 641046 Tamil Nadu India
| | - D. Sruthi
- Department of Biochemistry, Indian Institute of Science, Bengaluru, 560012 India
| | - K. R. Jinuraj
- OSPF-NIAS Drug Discovery Lab, NIAS, IISc Campus, Bengaluru, 560012 India
| | - Kajari Das
- Department of Biotechnology, College of Basic Science and Humanities, Odisha University of Agriculture and Technology, Bhubaneswar-3, Odisha India
| | - Sushma Dave
- Department of Applied Sciences, JIET, Jodhpur, Rajasthan India
| | - N. Muthulakshmi Andal
- Department of Chemistry, PSGR Krishnammal College for Women, Coimbatore, 641004 Tamil Nadu India
| | - Jayashankar Das
- Valnizen Healthcare, Vile Parle West, Mumbai, 400056 Maharashtra India
| |
Collapse
|
15
|
Efficacy of mucosal vaccination using a protozoan parasite as a vehicle for antigen delivery: IgG and neutralizing response after rectal administration of LeCoVax-2, a candidate vaccine against COVID-19. Pharmacol Res 2022; 186:106546. [PMCID: PMC9633108 DOI: 10.1016/j.phrs.2022.106546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/02/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022]
|
16
|
Tans R, Dey S, Dey NS, Cao JH, Paul PS, Calder G, O’Toole P, Kaye PM, Heeren RMA. Mass spectrometry imaging identifies altered hepatic lipid signatures during experimental Leishmania donovani infection. Front Immunol 2022; 13:862104. [PMID: 36003389 PMCID: PMC9394181 DOI: 10.3389/fimmu.2022.862104] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction Spatial analysis of lipids in inflammatory microenvironments is key to understand the pathogenesis of infectious disease. Granulomatous inflammation is a hallmark of leishmaniasis and changes in host and parasite lipid metabolism have been observed at the bulk tissue level in various infection models. Here, mass spectrometry imaging (MSI) is applied to spatially map hepatic lipid composition following infection with Leishmania donovani, an experimental mouse model of visceral leishmaniasis. Methods Livers from naïve and L. donovani-infected C57BL/6 mice were harvested at 14- and 20-days post-infection (n=5 per time point). 12 µm transverse sections were cut and covered with norhamane, prior to lipid analysis using MALDI-MSI. MALDI-MSI was performed in negative mode on a Rapiflex (Bruker Daltonics) at 5 and 50 µm spatial resolution and data-dependent analysis (DDA) on an Orbitrap-Elite (Thermo-Scientific) at 50 µm spatial resolution for structural identification analysis of lipids. Results Aberrant lipid abundances were observed in a heterogeneous distribution across infected mouse livers compared to naïve mouse liver. Distinctive localized correlated lipid masses were found in granulomas and surrounding parenchymal tissue. Structural identification revealed 40 different lipids common to naïve and d14/d20 infected mouse livers, whereas 15 identified lipids were only detected in infected mouse livers. For pathology-guided MSI imaging, we deduced lipids from manually annotated granulomatous and parenchyma regions of interests (ROIs), identifying 34 lipids that showed significantly different intensities between parenchyma and granulomas across all infected livers. Discussion Our results identify specific lipids that spatially correlate to the major histopathological feature of Leishmania donovani infection in the liver, viz. hepatic granulomas. In addition, we identified a three-fold increase in the number of unique phosphatidylglycerols (PGs) in infected liver tissue and provide direct evidence that arachidonic acid-containing phospholipids are localized with hepatic granulomas. These phospholipids may serve as important precursors for downstream oxylipin generation with consequences for the regulation of the inflammatory cascade. This study provides the first description of the use of MSI to define spatial-temporal lipid changes at local sites of infection induced by Leishmania donovani in mice.
Collapse
Affiliation(s)
- Roel Tans
- Maastricht MultiModal Molecular Imaging (M4I) Institute, Division of Imaging Mass Spectrometry, Maastricht University, Maastricht, Netherlands
| | - Shoumit Dey
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Nidhi Sharma Dey
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Jian-Hua Cao
- Maastricht MultiModal Molecular Imaging (M4I) Institute, Division of Imaging Mass Spectrometry, Maastricht University, Maastricht, Netherlands
| | - Prasanjit S. Paul
- Maastricht MultiModal Molecular Imaging (M4I) Institute, Division of Imaging Mass Spectrometry, Maastricht University, Maastricht, Netherlands
| | - Grant Calder
- Department of Biology, University of York, York, United Kingdom
| | - Peter O’Toole
- Department of Biology, University of York, York, United Kingdom
| | - Paul M. Kaye
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
- *Correspondence: Paul M. Kaye, ; Ron M. A. Heeren,
| | - Ron M. A. Heeren
- Maastricht MultiModal Molecular Imaging (M4I) Institute, Division of Imaging Mass Spectrometry, Maastricht University, Maastricht, Netherlands
- *Correspondence: Paul M. Kaye, ; Ron M. A. Heeren,
| |
Collapse
|
17
|
Dirkx L, Hendrickx S, Merlot M, Bulté D, Starick M, Elst J, Bafica A, Ebo DG, Maes L, Van Weyenbergh J, Caljon G. Long-term hematopoietic stem cells as a parasite niche during treatment failure in visceral leishmaniasis. Commun Biol 2022; 5:626. [PMID: 35752645 PMCID: PMC9233693 DOI: 10.1038/s42003-022-03591-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/14/2022] [Indexed: 01/20/2023] Open
Abstract
Given the discontinuation of various first-line drugs for visceral leishmaniasis (VL), large-scale in vivo drug screening, establishment of a relapse model in rodents, immunophenotyping, and transcriptomics were combined to study persistent infections and therapeutic failure. Double bioluminescent/fluorescent Leishmania infantum and L. donovani reporter lines enabled the identification of long-term hematopoietic stem cells (LT-HSC) as a niche in the bone marrow with remarkably high parasite burdens, a feature confirmed for human hematopoietic stem cells (hHSPC). LT-HSC are more tolerant to antileishmanial drug action and serve as source of relapse. A unique transcriptional ’StemLeish’ signature in these cells was defined by upregulated TNF/NF-κB and RGS1/TGF-β/SMAD/SKIL signaling, and a downregulated oxidative burst. Cross-species analyses demonstrated significant overlap with human VL and HIV co-infected blood transcriptomes. In summary, the identification of LT-HSC as a drug- and oxidative stress-resistant niche, undergoing a conserved transcriptional reprogramming underlying Leishmania persistence and treatment failure, may open therapeutic avenues for leishmaniasis. Long-term hematopoietic stem cells may act as protective niches for the Leishmania parasite, potentially contributing to treatment failure in cases of visceral leishmaniasis.
Collapse
Affiliation(s)
- Laura Dirkx
- Laboratory of Microbiology, Parasitology, and Hygiene (LMPH), Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Sarah Hendrickx
- Laboratory of Microbiology, Parasitology, and Hygiene (LMPH), Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Margot Merlot
- Laboratory of Microbiology, Parasitology, and Hygiene (LMPH), Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Dimitri Bulté
- Laboratory of Microbiology, Parasitology, and Hygiene (LMPH), Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Marick Starick
- Clinical and Epidemiological Virology, Department of Microbiology, Immunology, and Transplantation, Rega Institute of Medical Research, KU Leuven, Leuven, Belgium.,Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology Federal University of Santa Catarina, Florianopolis, Brazil
| | - Jessy Elst
- Department of Immunology-Allergology-Rheumatology, Faculty of Medicine and Health Science and the Infla-Med Centre of Excellence, University of Antwerp, Antwerp University Hospital, Antwerp, Belgium
| | - André Bafica
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology Federal University of Santa Catarina, Florianopolis, Brazil
| | - Didier G Ebo
- Department of Immunology-Allergology-Rheumatology, Faculty of Medicine and Health Science and the Infla-Med Centre of Excellence, University of Antwerp, Antwerp University Hospital, Antwerp, Belgium
| | - Louis Maes
- Laboratory of Microbiology, Parasitology, and Hygiene (LMPH), Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Johan Van Weyenbergh
- Clinical and Epidemiological Virology, Department of Microbiology, Immunology, and Transplantation, Rega Institute of Medical Research, KU Leuven, Leuven, Belgium
| | - Guy Caljon
- Laboratory of Microbiology, Parasitology, and Hygiene (LMPH), Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
18
|
Leishmania tarentolae as an Antigen Delivery Platform: Dendritic Cell Maturation after Infection with a Clone Engineered to Express the SARS-CoV-2 Spike Protein. Vaccines (Basel) 2022; 10:vaccines10050803. [PMID: 35632559 PMCID: PMC9144667 DOI: 10.3390/vaccines10050803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/05/2022] [Accepted: 05/18/2022] [Indexed: 01/18/2023] Open
Abstract
Background: Protozoa of the genus Leishmania are characterized by their capacity to target macrophages and Dendritic Cells (DCs). These microorganisms could thus be exploited for the delivery of antigens to immune cells. Leishmania tarentolae is regarded as a non-pathogenic species; it was previously used as a biofactory for protein production and has been considered as a candidate vaccine or as an antigen delivery platform. However, results on the type of immune polarization determined by L. tarentolae are still inconclusive. Methods: DCs were derived from human monocytes and exposed to live L. tarentolae, using both the non-engineered P10 strain, and the same strain engineered for expression of the spike protein from SARS-CoV-2. We then determined: (i) parasite internalization in the DCs; and (ii) the capacity of the assayed strains to activate DCs and the type of immune polarization. Results: Protozoan parasites from both strains were effectively engulfed by DCs, which displayed a full pattern of maturation, in terms of MHC class II and costimulatory molecule expression. In addition, after parasite infection, a limited release of Th1 cytokines was observed. Conclusions: Our results indicate that L. tarentolae could be used as a vehicle for antigen delivery to DCs and to induce the maturation of these cells. The limited cytokine release suggests L. tarentolae as a neutral vaccine vehicle that could be administered in association with appropriate immune-modulating molecules.
Collapse
|
19
|
Volpedo G, Pacheco-Fernandez T, Holcomb EA, Zhang WW, Lypaczewski P, Cox B, Fultz R, Mishan C, Verma C, Huston RH, Wharton AR, Dey R, Karmakar S, Oghumu S, Hamano S, Gannavaram S, Nakhasi HL, Matlashewski G, Satoskar AR. Centrin-deficient Leishmania mexicana confers protection against New World cutaneous leishmaniasis. NPJ Vaccines 2022; 7:32. [PMID: 35236861 PMCID: PMC8891280 DOI: 10.1038/s41541-022-00449-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 01/27/2022] [Indexed: 01/01/2023] Open
Abstract
Leishmaniasis is a neglected protozoan disease affecting over 12 million people globally with no approved vaccines for human use. New World cutaneous leishmaniasis (CL) caused by L. mexicana is characterized by the development of chronic non-healing skin lesions. Using the CRISPR/Cas9 technique, we have generated live attenuated centrin knockout L. mexicana (LmexCen-/-) parasites. Centrin is a cytoskeletal protein important for cellular division in eukaryotes and, in Leishmania, is required only for intracellular amastigote replication. We have investigated the safety and immunogenicity characteristics of LmexCen-/- parasites by evaluating their survival and the cytokine production in bone-marrow-derived macrophages (BMDMs) and dendritic cells (BMDCs) in vitro. Our data shows that LmexCen-/- amastigotes present a growth defect, which results in significantly lower parasitic burdens and increased protective cytokine production in infected BMDMs and BMDCs, compared to the wild type (WT) parasites. We have also determined the safety and efficacy of LmexCen-/- in vivo using experimental murine models of L. mexicana. We demonstrate that LmexCen-/- parasites are safe and do not cause lesions in susceptible mouse models. Immunization with LmexCen-/- is also efficacious against challenge with WT L. mexicana parasites in genetically different BALB/c and C57BL/6 mouse models. Vaccinated mice did not develop cutaneous lesions, displayed protective immunity, and showed significantly lower parasitic burdens at the infection site and draining lymph nodes compared to the control group. Overall, we demonstrate that LmexCen-/- parasites are safe and efficacious against New World cutaneous leishmaniasis in pre-clinical models.
Collapse
Affiliation(s)
- Greta Volpedo
- Department of Microbiology, The Ohio State University, Columbus, OH, 43210, USA.,Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Thalia Pacheco-Fernandez
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Erin A Holcomb
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Wen-Wei Zhang
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Patrick Lypaczewski
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Blake Cox
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Rebecca Fultz
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Chelsea Mishan
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Chaitenya Verma
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Ryan H Huston
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Abigail R Wharton
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Ranadhir Dey
- Division of Emerging and Transfusion Transmitted Diseases, CBER, FDA, Silver Spring, MD, USA
| | - Subir Karmakar
- Division of Emerging and Transfusion Transmitted Diseases, CBER, FDA, Silver Spring, MD, USA
| | - Steve Oghumu
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Shinjiro Hamano
- Department of Parasitology, Institute of Tropical Medicine (NEKKEN), The Joint Usage/Research Center on Tropical Disease, Nagasaki University, Nagasaki University Graduate School of Biomedical Sciences Doctoral Leadership Program, Nagasaki, Japan
| | - Sreenivas Gannavaram
- Division of Emerging and Transfusion Transmitted Diseases, CBER, FDA, Silver Spring, MD, USA
| | - Hira L Nakhasi
- Division of Emerging and Transfusion Transmitted Diseases, CBER, FDA, Silver Spring, MD, USA.
| | - Greg Matlashewski
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada.
| | - Abhay R Satoskar
- Department of Microbiology, The Ohio State University, Columbus, OH, 43210, USA. .,Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
20
|
The Ecto-5
′
nucleotidase/CD73 Mediates Leishmania amazonensis Survival in Macrophages. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9928362. [PMID: 35187176 PMCID: PMC8856795 DOI: 10.1155/2022/9928362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 11/30/2021] [Accepted: 12/28/2021] [Indexed: 11/23/2022]
Abstract
Endogenous nucleotides produced by various group of cells under inflammatory conditions act as potential danger signals in vivo. Extracellularly released nucleotides such as ATP are rapidly hydrolyzed to adenosine by the coordinated ectonucleotidase activities of CD39 and CD73. Leishmania is an obligate intracellular parasite of macrophages and capable of modulating host immune response in order to survive and multiply within host cells. In this study, the activity of CD73 induced by Leishmania amazonensis in infected macrophages has been investigated and correlated with parasite survival and infection in vitro. For this, the expression of CD39 and CD73, by flow cytometry, in murine peritoneal macrophages infected with metacyclic promastigotes of L. amazonensis has been analyzed. Our results showed that L. amazonensis-infected macrophages, unlike LPS-treated macrophages, increased CD73 expression. It was also noted that when CD73 enzymatic activity was blocked by α, β-methyleneadenosine 5′-diphosphate sodium salt (APCP), macrophage parasitism was significantly decreased. Interestingly, these effects were not associated with the production of TNF-α, IL-10, or nitric oxide (NO). Together, these data demonstrate that L. amazonensis induces a regulatory phenotype in macrophages, which by activating the CD39/CD73 pathway allows parasite survival through the action of immunomodulatory adenosine receptors.
Collapse
|
21
|
Sanz CR, Miró G, Sevane N, Reyes-Palomares A, Dunner S. Modulation of Host Immune Response during Leishmania infantum Natural Infection: A Whole-Transcriptome Analysis of the Popliteal Lymph Nodes in Dogs. Front Immunol 2022; 12:794627. [PMID: 35058931 PMCID: PMC8763708 DOI: 10.3389/fimmu.2021.794627] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/10/2021] [Indexed: 12/21/2022] Open
Abstract
Leishmania infantum, the etiological agent of canine leishmaniosis (CanL) in Europe, was responsible of the largest outbreak of human leishmaniosis in Spain. The parasite infects and survives within myeloid lineage cells, causing a potentially fatal disease if left untreated. The only treatment option relies on chemotherapy, although immunotherapy strategies are being considered as novel approaches to prevent progression of the disease. To this aim, a deeper characterization of the molecular mechanisms behind the immunopathogenesis of leishmaniosis is necessary. Thus, we evaluated, for the first time, the host immune response during L. infantum infection through transcriptome sequencing of the popliteal lymph nodes aspirates of dogs with CanL. Differential expression and weighted gene co-expression network analyses were performed, resulting in the identification of 5,461 differentially expressed genes (DEGs) and four key modules in sick dogs, compared to controls. As expected, defense response was the highest enriched biological process in the DEGs, with six genes related to immune response against pathogens (CHI3L1, SLPI, ACOD1, CCL5, MPO, BPI) included among the ten most expressed genes; and two of the key co-expression modules were associated with regulation of immune response, which also positively correlated with clinical stage and blood monocyte concentration. In particular, sick dogs displayed significant changes in the expression of Th1, Th2, Th17 and Tr1 cytokines (e. g. TNF-α, IFN-γ, IL-21, IL-17, IL-15), markers of T cell and NK cell exhaustion (e. g. LAG3, CD244, Blimp-1, JUN), and B cell, monocyte and macrophage disrupted functionality (e. g. CD40LG, MAPK4, IL-1R, NLRP3, BCMA). In addition, we found an overexpression of XBP1 and some other genes involved in endoplasmic reticulum stress and the IRE1 branch of the unfolded protein response, as well as one co-expression module associated with these processes, which could be induced by L. infantum to prevent host cell apoptosis and modulate inflammation-induced lymphangiogenesis at lymph nodes. Moreover, 21 lncRNAs were differentially expressed in sick dogs, and one key co-expression module was associated with chromatin organization, suggesting that epigenetic mechanisms could also contribute to dampening host immune response during natural L. infantum infection in the lymph nodes of dogs suffering from clinical leishmaniosis.
Collapse
Affiliation(s)
- Carolina R Sanz
- Animal Health Department, Veterinary Faculty, Complutense University of Madrid, Madrid, Spain
| | - Guadalupe Miró
- Animal Health Department, Veterinary Faculty, Complutense University of Madrid, Madrid, Spain
| | - Natalia Sevane
- Department of Animal Production, Veterinary Faculty, Complutense University of Madrid, Madrid, Spain
| | - Armando Reyes-Palomares
- Department of Biochemistry and Molecular Biology, Complutense University of Madrid, Madrid, Spain
| | - Susana Dunner
- Department of Animal Production, Veterinary Faculty, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
22
|
Jara M, Barrett M, Maes I, Regnault C, Imamura H, Domagalska MA, Dujardin JC. Transcriptional Shift and Metabolic Adaptations during Leishmania Quiescence Using Stationary Phase and Drug Pressure as Models. Microorganisms 2022; 10:97. [PMID: 35056546 PMCID: PMC8781126 DOI: 10.3390/microorganisms10010097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/28/2021] [Accepted: 12/30/2021] [Indexed: 11/16/2022] Open
Abstract
Microorganisms can adopt a quiescent physiological condition which acts as a survival strategy under unfavorable conditions. Quiescent cells are characterized by slow or non-proliferation and a deep downregulation of processes related to biosynthesis. Although quiescence has been described mostly in bacteria, this survival skill is widespread, including in eukaryotic microorganisms. In Leishmania, a digenetic parasitic protozoan that causes a major infectious disease, quiescence has been demonstrated, but the molecular and metabolic features enabling its maintenance are unknown. Here, we quantified the transcriptome and metabolome of Leishmania promastigotes and amastigotes where quiescence was induced in vitro either, through drug pressure or by stationary phase. Quiescent cells have a global and coordinated reduction in overall transcription, with levels dropping to as low as 0.4% of those in proliferating cells. However, a subset of transcripts did not follow this trend and were relatively upregulated in quiescent populations, including those encoding membrane components, such as amastins and GP63, or processes like autophagy. The metabolome followed a similar trend of overall downregulation albeit to a lesser magnitude than the transcriptome. It is noteworthy that among the commonly upregulated metabolites were those involved in carbon sources as an alternative to glucose. This first integrated two omics layers afford novel insight into cell regulation and show commonly modulated features across stimuli and stages.
Collapse
Affiliation(s)
- Marlene Jara
- Molecular Parasitology Unit, Institute of Tropical Medicine Antwerp, 2000 Antwerp, Belgium; (I.M.); (M.A.D.)
| | - Michael Barrett
- Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK; (M.B.); (C.R.)
- Glasgow Polyomics, Wolfson Wohl Cancer Research Centre, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Ilse Maes
- Molecular Parasitology Unit, Institute of Tropical Medicine Antwerp, 2000 Antwerp, Belgium; (I.M.); (M.A.D.)
| | - Clement Regnault
- Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK; (M.B.); (C.R.)
- Glasgow Polyomics, Wolfson Wohl Cancer Research Centre, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Hideo Imamura
- Centre for Medical Genetics, Universitair Ziekenhuis Brussel, 1090 Brussels, Belgium;
| | - Malgorzata Anna Domagalska
- Molecular Parasitology Unit, Institute of Tropical Medicine Antwerp, 2000 Antwerp, Belgium; (I.M.); (M.A.D.)
| | - Jean-Claude Dujardin
- Molecular Parasitology Unit, Institute of Tropical Medicine Antwerp, 2000 Antwerp, Belgium; (I.M.); (M.A.D.)
- Department of Biomedical Sciences, University of Antwerp, 2000 Antwerp, Belgium
| |
Collapse
|
23
|
Doehl JSP, Ashwin H, Brown N, Romano A, Carmichael S, Pitchford JW, Kaye PM. Spatial Point Pattern Analysis Identifies Mechanisms Shaping the Skin Parasite Landscape in Leishmania donovani Infection. Front Immunol 2021; 12:795554. [PMID: 34975901 PMCID: PMC8716623 DOI: 10.3389/fimmu.2021.795554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 11/23/2021] [Indexed: 12/15/2022] Open
Abstract
Increasing evidence suggests that in hosts infected with parasites of the Leishmania donovani complex, transmission of infection to the sand fly vector is linked to parasite repositories in the host skin. However, a detailed understanding of the dispersal (the mechanism of spread) and dispersion (the observed state of spread) of these obligatory-intracellular parasites and their host phagocytes in the skin is lacking. Using endogenously fluorescent parasites as a proxy, we apply image analysis combined with spatial point pattern models borrowed from ecology to characterize dispersion of parasitized myeloid cells (including ManR+ and CD11c+ cells) and predict dispersal mechanisms in a previously described immunodeficient model of L. donovani infection. Our results suggest that after initial seeding of infection in the skin, heavily parasite-infected myeloid cells are found in patches that resemble innate granulomas. Spread of parasites from these initial patches subsequently occurs through infection of recruited myeloid cells, ultimately leading to self-propagating networks of patch clusters. This combination of imaging and ecological pattern analysis to identify mechanisms driving the skin parasite landscape offers new perspectives on myeloid cell behavior following parasitism by L. donovani and may also be applicable to elucidating the behavior of other intracellular tissue-resident pathogens and their host cells.
Collapse
MESH Headings
- Animals
- CD11 Antigens/metabolism
- Cluster Analysis
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Disease Models, Animal
- Host-Parasite Interactions
- Image Processing, Computer-Assisted
- Insect Vectors/parasitology
- Leishmania donovani/immunology
- Leishmania donovani/pathogenicity
- Leishmaniasis, Visceral/immunology
- Leishmaniasis, Visceral/metabolism
- Leishmaniasis, Visceral/parasitology
- Leishmaniasis, Visceral/transmission
- Mannose Receptor/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Microscopy, Confocal
- Microscopy, Fluorescence
- Models, Theoretical
- Myeloid Cells/immunology
- Myeloid Cells/metabolism
- Myeloid Cells/parasitology
- Phlebotomus/parasitology
- Skin/immunology
- Skin/metabolism
- Skin/parasitology
- Spatial Analysis
- Mice
Collapse
Affiliation(s)
- Johannes S. P. Doehl
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Helen Ashwin
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Najmeeyah Brown
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Audrey Romano
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Samuel Carmichael
- Departments of Biology and Mathematics, University of York, York, United Kingdom
| | - Jon W. Pitchford
- Departments of Biology and Mathematics, University of York, York, United Kingdom
| | - Paul M. Kaye
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| |
Collapse
|
24
|
Tandon S, Muthuswami R, Madhubala R. Role of two aminoacyl-tRNA synthetase associated proteins (Endothelial Monocyte Activating Polypeptides 1 and 2) of Leishmania donovani in chemotaxis of human monocytes. Acta Trop 2021; 224:106128. [PMID: 34509454 DOI: 10.1016/j.actatropica.2021.106128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 09/02/2021] [Accepted: 09/02/2021] [Indexed: 11/26/2022]
Abstract
Visceral leishmaniasis is caused by the protozoan parasite Leishmania donovani. It is a fatal form of leishmaniasis prevalent in Indian subcontinent. Since there are no human licensed vaccines available for leishmaniasis, chemotherapeutic drugs remain the only means for combating parasitic infections. We have earlier identified a total of 26 amino-acyl tRNA synthetases (aaRS) along with five stand-alone editing domains and two aaRS-associated proteins in Leishmania donovani. In addition to their canonical role of tRNA aminoacylation, aaRS have been involved in novel functions by acquiring novel domains during evolution. The aaRS-associated proteins have been reported to be analogous to a human cytokine, EMAP II, as they possess a modified version of the heptapeptide motif responsible for the cytokine activity. In this manuscript, we report the characterization of two L. donovani aminoacyl-tRNA synthetase associated proteins which showed a human chemokine like activity. Both the proteins, L. donovani EMAP-1 and EMAP-2, possess a modified form of the heptapeptide motif, which is responsible for cytokine activity in human EMAP-2. LdEMAP-1 and LdEMAP-2 were cloned, expressed, and purified. Both LdEMAP-1 and LdEMAP-2 proteins in the promastigote stage were found to be localized in cytoplasm as confirmed by immunofluorescence. In case of L. donovani infected human THP-1 derived macrophages, secretion of LdEMAP-1 and LdEMAP-2 proteins in the cytosol of the macrophages was observed. The role of LdEMAP-1 and LdEMAP-2 in the aminoacylation of rLdTyrRS was also tested and LdEMAP-2 but not LdEMAP-1 increased the rate of aminoacylation of tyrosyl tRNA synthetase (rLdTyrRS). L. donovani EMAP-1 and EMAP-2 proteins managed to exhibit the capability of attracting human origin cells as determined by chemotaxis assay, and also were able to induce the secretion of cytokines from macrophages like their human counterpart (EMAP II). Our working hypothesis is that both of these proteins might be involved in helping the parasite to establish the infection within the host.
Collapse
|
25
|
Kolářová I, Valigurová A. Hide-and-Seek: A Game Played between Parasitic Protists and Their Hosts. Microorganisms 2021; 9:2434. [PMID: 34946036 PMCID: PMC8707157 DOI: 10.3390/microorganisms9122434] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/17/2021] [Accepted: 11/20/2021] [Indexed: 11/17/2022] Open
Abstract
After invading the host organism, a battle occurs between the parasitic protists and the host's immune system, the result of which determines not only whether and how well the host survives and recovers, but also the fate of the parasite itself. The exact weaponry of this battle depends, among others, on the parasite localisation. While some parasitic protists do not invade the host cell at all (extracellular parasites), others have developed successful intracellular lifestyles (intracellular parasites) or attack only the surface of the host cell (epicellular parasites). Epicellular and intracellular protist parasites have developed various mechanisms to hijack host cell functions to escape cellular defences and immune responses, and, finally, to gain access to host nutrients. They use various evasion tactics to secure the tight contact with the host cell and the direct nutrient supply. This review focuses on the adaptations and evasion strategies of parasitic protists on the example of two very successful parasites of medical significance, Cryptosporidium and Leishmania, while discussing different localisation (epicellular vs. intracellular) with respect to the host cell.
Collapse
Affiliation(s)
- Iva Kolářová
- Laboratory of Vector Biology, Department of Parasitology, Faculty of Science, Charles University, Albertov 6, 128 44 Prague, Czech Republic
| | - Andrea Valigurová
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, 611 37 Brno, Czech Republic
| |
Collapse
|
26
|
Barroso DH, Nóbrega ODT, de Araújo CN, Freire GSM, Martins SS, Rodrigues BC, Gomes CM, Sampaio RNR. The Presence of Leishmania braziliensis DNA in the Nasal Mucosa of Cutaneous Leishmaniasis Patients and the Search for Possible Clinical and Immunological Patterns of Disease Progression: A Cross Sectional Study. Front Cell Infect Microbiol 2021; 11:744163. [PMID: 34722337 PMCID: PMC8551912 DOI: 10.3389/fcimb.2021.744163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/28/2021] [Indexed: 11/15/2022] Open
Abstract
Leishmania braziliensis is the most important causal agent of American tegumentary leishmaniasis (ATL), and 3 to 5% of patients develop mucosal lesions. The mechanisms related to parasite and host immune interactions and the parasite life cycle that lead to dissemination to the mucosa are poorly understood. We aimed to detect L. braziliensis DNA in the nasal mucosa of cutaneous leishmaniasis (CL) patients with early mucous dissemination and to relate those findings to specific inflammatory responses. Nasal swabs were collected from patients with the cutaneous form of ATL. L. braziliensis DNA was investigated using TaqMan-based real-time PCR. The levels of serum cytokines (IL-12, IL-6, TNF-α, IL-10, IL-1β and IL-8) were measured by a multiplex cytometric array. A Poisson regression model was used to test prevalence ratios (PRs) and multivariate interactions of clinical and laboratory characteristics. Of the 79 CL patients, 24 (30%) had L. braziliensis DNA in the nasal mucosa. In the multivariate model, parasite DNA presence in mucosa was associated with a reduction in IL-12 levels (PR = 0.440; p=0.034), increased IL-6 levels (PR = 1.001; p=0.002) and a higher number of affected body segments (PR = 1.65; p<0.001). In this study, we observed a higher rate of early dissemination to the nasal mucosa than what was previously described. We suggest that an enhanced Th1 profile characterized by higher IL-12 is important for preventing dissemination of L. braziliensis to the mucosa. Further evaluation of parasite-related interactions with the host immunological response is necessary to elucidate the dissemination mechanisms of Leishmania.
Collapse
Affiliation(s)
- Daniel Holanda Barroso
- Programa de Pós-Graduação em Ciências Médicas, Faculdade de Medicina, Universidade de Brasília (UnB), Brasília, Brazil.,Hospital Universitário de Brasília, Universidade de Brasília, Brasília, Brazil.,Laboratório de Dermatomicologia da Faculdade de Medicina, Universidade de Brasília, Brasília, Brazil
| | - Otávio de Toledo Nóbrega
- Programa de Pós-Graduação em Ciências Médicas, Faculdade de Medicina, Universidade de Brasília (UnB), Brasília, Brazil.,Pós-Graduação de Ciências da Saúde da Faculdade de Ciências Saúde, Universidade de Brasília, Brasília, Brazil
| | - Carla Nunes de Araújo
- Programa de Pós-Graduação em Ciências Médicas, Faculdade de Medicina, Universidade de Brasília (UnB), Brasília, Brazil
| | | | - Sofia Sales Martins
- Hospital Universitário de Brasília, Universidade de Brasília, Brasília, Brazil.,Pós-Graduação de Ciências da Saúde da Faculdade de Ciências Saúde, Universidade de Brasília, Brasília, Brazil
| | - Bruna Côrtes Rodrigues
- Programa de Pós-Graduação em Ciências Médicas, Faculdade de Medicina, Universidade de Brasília (UnB), Brasília, Brazil.,Hospital Universitário de Brasília, Universidade de Brasília, Brasília, Brazil
| | - Ciro Martins Gomes
- Programa de Pós-Graduação em Ciências Médicas, Faculdade de Medicina, Universidade de Brasília (UnB), Brasília, Brazil.,Hospital Universitário de Brasília, Universidade de Brasília, Brasília, Brazil.,Laboratório de Dermatomicologia da Faculdade de Medicina, Universidade de Brasília, Brasília, Brazil.,Programa de Pós-Graduação em Medicina Tropical, Faculdade de Medicina, Universidade de Brasília (UnB), Brasília, Brazil
| | - Raimunda Nonata Ribeiro Sampaio
- Programa de Pós-Graduação em Ciências Médicas, Faculdade de Medicina, Universidade de Brasília (UnB), Brasília, Brazil.,Hospital Universitário de Brasília, Universidade de Brasília, Brasília, Brazil.,Laboratório de Dermatomicologia da Faculdade de Medicina, Universidade de Brasília, Brasília, Brazil.,Pós-Graduação de Ciências da Saúde da Faculdade de Ciências Saúde, Universidade de Brasília, Brasília, Brazil
| |
Collapse
|
27
|
Volpedo G, Pacheco-Fernandez T, Bhattacharya P, Oljuskin T, Dey R, Gannavaram S, Satoskar AR, Nakhasi HL. Determinants of Innate Immunity in Visceral Leishmaniasis and Their Implication in Vaccine Development. Front Immunol 2021; 12:748325. [PMID: 34712235 PMCID: PMC8546207 DOI: 10.3389/fimmu.2021.748325] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 09/24/2021] [Indexed: 12/22/2022] Open
Abstract
Leishmaniasis is endemic to the tropical and subtropical regions of the world and is transmitted by the bite of an infected sand fly. The multifaceted interactions between Leishmania, the host innate immune cells, and the adaptive immunity determine the severity of pathogenesis and disease development. Leishmania parasites establish a chronic infection by subversion and attenuation of the microbicidal functions of phagocytic innate immune cells such as neutrophils, macrophages and dendritic cells (DCs). Other innate cells such as inflammatory monocytes, mast cells and NK cells, also contribute to resistance and/or susceptibility to Leishmania infection. In addition to the cytokine/chemokine signals from the innate immune cells, recent studies identified the subtle shifts in the metabolic pathways of the innate cells that activate distinct immune signal cascades. The nexus between metabolic pathways, epigenetic reprogramming and the immune signaling cascades that drive the divergent innate immune responses, remains to be fully understood in Leishmania pathogenesis. Further, development of safe and efficacious vaccines against Leishmaniasis requires a broader understanding of the early interactions between the parasites and innate immune cells. In this review we focus on the current understanding of the specific role of innate immune cells, the metabolomic and epigenetic reprogramming and immune regulation that occurs during visceral leishmaniasis, and the strategies used by the parasite to evade and modulate host immunity. We highlight how such pathways could be exploited in the development of safe and efficacious Leishmania vaccines.
Collapse
Affiliation(s)
- Greta Volpedo
- Departments of Pathology and Microbiology, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Thalia Pacheco-Fernandez
- Departments of Pathology and Microbiology, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Parna Bhattacharya
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Timur Oljuskin
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Ranadhir Dey
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Sreenivas Gannavaram
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Abhay R Satoskar
- Departments of Pathology and Microbiology, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Hira L Nakhasi
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| |
Collapse
|
28
|
Liang C, Zhao Y, Chen C, Huang S, Deng T, Zeng X, Tan J, Zha X, Chen S, Li Y. Higher TOX Genes Expression Is Associated With Poor Overall Survival for Patients With Acute Myeloid Leukemia. Front Oncol 2021; 11:740642. [PMID: 34692519 PMCID: PMC8532529 DOI: 10.3389/fonc.2021.740642] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 09/20/2021] [Indexed: 12/11/2022] Open
Abstract
Thymocyte selection-associated HMG box (TOX) is a transcription factor that belongs to the high mobility group box (HMG-box) superfamily, which includes four subfamily members: TOX, TOX2, TOX3, and TOX4. TOX is related to the formation of multiple malignancies and contributes to CD8+ T cell exhaustion in solid tumors. However, little is known about the role of TOX genes in hematological malignancies. In this study, we explored the prognostic value of TOX genes from 40 patients with de novo acute myeloid leukemia (AML) by quantitative real-time PCR (qRT-PCR) in a training cohort and validated the results using transcriptome data from 167 de novo AML patients from the Cancer Genome Atlas (TCGA) database. In the training cohort, higher expression of TOX and TOX4 was detected in the AML samples, whereas lower TOX3 expression was found. Moreover, both the training and validation results indicated that higher TOX2, TOX3, and TOX4 expression of AML patients (3-year OS: 0% vs. 37%, P = 0.036; 3-year OS: 4% vs. 61%, P < 0.001; 3-year OS: 0% vs. 32%, P = 0.010) and the AML patients with highly co-expressed TOX, TOX2, TOX4 genes (3-year OS: 0% vs. 25% vs. 75%, P = 0.001) were associated with poor overall survival (OS). Interestingly, TOX2 was positively correlated with CTLA-4, PD-1, TIGIT, and PDL-2 (rs = 0.43, P = 0.006; rs = 0.43, P = 0.006; rs = 0.56, P < 0.001; rs = 0.54, P < 0.001). In conclusion, higher expression of TOX genes was associated with poor OS for AML patients, which was related to the up-regulation of immune checkpoint genes. These data might provide novel predictors for AML outcome and direction for further investigation of the possibility of using TOX genes in novel targeted therapies for AML.
Collapse
Affiliation(s)
- Chaofeng Liang
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Yujie Zhao
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Cunte Chen
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Shuxin Huang
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Tairan Deng
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Xiangbo Zeng
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Jiaxiong Tan
- Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Xianfeng Zha
- Department of Clinical Laboratory, First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Shaohua Chen
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Yangqiu Li
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| |
Collapse
|
29
|
Hohman LS, Mou Z, Carneiro MB, Ferland G, Kratofil RM, Kubes P, Uzonna JE, Peters NC. Protective CD4+ Th1 cell-mediated immunity is reliant upon execution of effector function prior to the establishment of the pathogen niche. PLoS Pathog 2021; 17:e1009944. [PMID: 34543348 PMCID: PMC8483310 DOI: 10.1371/journal.ppat.1009944] [Citation(s) in RCA: 8] [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: 05/11/2021] [Revised: 09/30/2021] [Accepted: 09/07/2021] [Indexed: 11/30/2022] Open
Abstract
Intracellular infection with the parasite Leishmania major features a state of concomitant immunity in which CD4+ T helper 1 (Th1) cell-mediated immunity against reinfection coincides with a chronic but sub-clinical primary infection. In this setting, the rapidity of the Th1 response at a secondary site of challenge in the skin represents the best correlate of parasite elimination and has been associated with a reversal in Leishmania-mediated modulation of monocytic host cells. Remarkably, the degree to which Th1 cells are absolutely reliant upon the time at which they interact with infected monocytes to mediate their protective effect has not been defined. In the present work, we report that CXCR3-dependent recruitment of Ly6C+ Th1 effector (Th1EFF) cells is indispensable for concomitant immunity and acute (<4 days post-infection) Th1EFF cell-phagocyte interactions are critical to prevent the establishment of a permissive pathogen niche, as evidenced by altered recruitment, gene expression and functional capacity of innate and adaptive immune cells at the site of secondary challenge. Surprisingly, provision of Th1EFF cells after establishment of the pathogen niche, even when Th1 cells were provided in large quantities, abrogated protection, Th1EFF cell accumulation and IFN-γ production, and iNOS production by inflammatory monocytes. These findings indicate that protective Th1 immunity is critically dependent on activation of permissive phagocytic host cells by preactivated Th1EFF cells at the time of infection.
Collapse
Affiliation(s)
- Leah S. Hohman
- Snyder Institute for Chronic Diseases; Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, Calgary, Alberta, Canada
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine; University of Calgary, Calgary, Alberta, Canada
| | - Zhirong Mou
- Department of Immunology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Matheus B. Carneiro
- Snyder Institute for Chronic Diseases; Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, Calgary, Alberta, Canada
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine; University of Calgary, Calgary, Alberta, Canada
| | - Gabriel Ferland
- Snyder Institute for Chronic Diseases; Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, Calgary, Alberta, Canada
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine; University of Calgary, Calgary, Alberta, Canada
| | - Rachel M. Kratofil
- Snyder Institute for Chronic Diseases; Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Paul Kubes
- Snyder Institute for Chronic Diseases; Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Jude E. Uzonna
- Department of Immunology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Nathan C. Peters
- Snyder Institute for Chronic Diseases; Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, Calgary, Alberta, Canada
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine; University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
30
|
Chen JY, Zhou JK, Pan W. Immunometabolism: Towards a Better Understanding the Mechanism of Parasitic Infection and Immunity. Front Immunol 2021; 12:661241. [PMID: 34122419 PMCID: PMC8191844 DOI: 10.3389/fimmu.2021.661241] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 05/13/2021] [Indexed: 12/26/2022] Open
Abstract
As a relatively successful pathogen, several parasites can establish long-term infection in host. This “harmonious symbiosis” status relies on the “precise” manipulation of host immunity and metabolism, however, the underlying mechanism is still largely elusive. Immunometabolism is an emerging crossed subject in recent years. It mainly discusses the regulatory mechanism of metabolic changes on reprogramming the key transcriptional and post-transcriptional events related to immune cell activation and effect, which provides a novel insight for understanding how parasites regulate the infection and immunity in hosts. The present study reviewed the current research progress on metabolic reprogramming mechanism exploited by parasites to modulate the function in various immune cells, highlighting the future exploitation of key metabolites or metabolic events to clarify the underlying mechanism of anti-parasite immunity and design novel intervention strategies against parasitic infection.
Collapse
Affiliation(s)
- Jing-Yue Chen
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China.,The First Clinical Medicine, Xuzhou Medical University, Xuzhou, China
| | - Ji-Kai Zhou
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China.,The First Clinical Medicine, Xuzhou Medical University, Xuzhou, China
| | - Wei Pan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
31
|
Elmahallawy EK, Alkhaldi AAM, Saleh AA. Host immune response against leishmaniasis and parasite persistence strategies: A review and assessment of recent research. Biomed Pharmacother 2021; 139:111671. [PMID: 33957562 DOI: 10.1016/j.biopha.2021.111671] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 04/18/2021] [Accepted: 04/24/2021] [Indexed: 12/13/2022] Open
Abstract
Leishmaniasis, a neglected parasitic disease caused by a unicellular protozoan of the genus Leishmania, is transmitted through the bite of a female sandfly. The disease remains a major public health problem and is linked to tropical and subtropical regions, with an endemic picture in several regions, including East Africa, the Mediterranean basin and South America. The different causative species display a diversity of clinical presentations; therefore, the immunological data on leishmaniasis are both scarce and controversial for the different forms and infecting species of the parasite. The present review highlights the main immune parameters associated with leishmaniasis that might contribute to a better understanding of the pathogenicity of the parasite and the clinical outcomes of the disease. Our aim was to provide a concise overview of the immunobiology of the disease and the factors that influence it, as this knowledge may be helpful in developing novel chemotherapeutic and vaccine strategies.
Collapse
Affiliation(s)
- Ehab Kotb Elmahallawy
- Department of Zoonoses, Faculty of Veterinary Medicine, Sohag University, Sohag 82524, Egypt.
| | | | - Amira A Saleh
- Department of Medical Parasitology, Faculty of Medicine, Zagazig University, Zgazig, Egypt
| |
Collapse
|
32
|
Mota CA, Oyama J, Souza Terron Monich MD, Brustolin AÁ, Perez de Souza JV, Murase LS, Ghiraldi Lopes LD, Silva Santos TD, Vieira Teixeira JJ, Verzignassi Silveira TG. Three decades of clinical trials on immunotherapy for human leishmaniases: a systematic review and meta-analysis. Immunotherapy 2021; 13:693-721. [PMID: 33853344 DOI: 10.2217/imt-2020-0184] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Aim: Current treatments for leishmaniases are not satisfactory, thus alternatives are needed. We searched for clinical trials with immunotherapeutic approaches for patients with leishmaniasis. Materials & methods: Out of 205 articles, 24 clinical trials were selected, and eight submitted to meta-analysis. Results: A reduction in healing time was observed in patients with tegumentary leishmaniasis treated with pentavalent antimony plus granulocyte-macrophage colony-stimulating factor, and therapeutic vaccines. Overall meta-analysis indicated that immunotherapy associated with the standard chemotherapy generated a significantly reduced risk of treatment failure than the pentavalent antimony alone (p = 0.03). Conclusion: Our review confirmed the efficacy of immunotherapies for the treatment of cutaneous and visceral leishmaniasis and highlighted the importance of clinical trials using immunotherapies for leishmaniases.
Collapse
Affiliation(s)
- Camila Alves Mota
- Graduate Program in Health Sciences, State University of Maringá, Maringá, Paraná, Brazil
| | - Jully Oyama
- Graduate Program in Bioscience & Physiopathology, State University of Maringá, Maringá, Paraná, Brazil
| | | | - Aline Ávila Brustolin
- Graduate Program in Health Sciences, State University of Maringá, Maringá, Paraná, Brazil
| | - João Vítor Perez de Souza
- Graduate Program in Bioscience & Physiopathology, State University of Maringá, Maringá, Paraná, Brazil
| | - Letícia Sayuri Murase
- Graduate Program in Health Sciences, State University of Maringá, Maringá, Paraná, Brazil
| | - Luciana Dias Ghiraldi Lopes
- Laboratory of Clinical Virology, Department of Clinical Analysis & Biomedicine, State University of Maringá, Maringá, Paraná, Brazil
| | - Thais da Silva Santos
- Graduate Program in Bioscience & Physiopathology, State University of Maringá, Maringá, Paraná, Brazil
| | - Jorge Juarez Vieira Teixeira
- Laboratory of Leishmaniases, Department of Clinical Analysis & Biomedicine, State University of Maringá, Maringá, Paraná, Brazil
| | - Thaís Gomes Verzignassi Silveira
- Laboratory of Leishmaniases, Department of Clinical Analysis & Biomedicine, State University of Maringá, Maringá, Paraná, Brazil
| |
Collapse
|
33
|
Pacheco-Fernandez T, Volpedo G, Gannavaram S, Bhattacharya P, Dey R, Satoskar A, Matlashewski G, Nakhasi HL. Revival of Leishmanization and Leishmanin. Front Cell Infect Microbiol 2021; 11:639801. [PMID: 33816344 PMCID: PMC8010169 DOI: 10.3389/fcimb.2021.639801] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 02/05/2021] [Indexed: 11/16/2022] Open
Abstract
Leishmaniasis includes a spectrum of diseases ranging from debilitating cutaneous to fatal visceral infections. This disease is caused by the parasitic protozoa of the genus Leishmania that is transmitted by infected sandflies. Over 1 billion people are at risk of leishmaniasis with an annual incidence of over 2 million cases throughout tropical and subtropical regions in close to 100 countries. Leishmaniasis is the only human parasitic disease where vaccination has been successful through a procedure known as leishmanization that has been widely used for decades in the Middle East. Leishmanization involved intradermal inoculation of live Leishmania major parasites resulting in a skin lesion that following natural healing provided protective immunity to re-infection. Leishmanization is however no longer practiced due to safety and ethical concerns that the lesions at the site of inoculation that can last for months in some people. New genome editing technologies involving CRISPR has now made it possible to engineer safer attenuated strains of Leishmania, which induce protective immunity making way for a second generation leishmanization that can enter into human trials. A major consideration will be how the test the efficacy of a vaccine in the midst of the visceral leishmaniasis elimination program. One solution will be to use the leishmanin skin test (LST) that was also used for decades to determine exposure and immunity to Leishmania. The LST involves injection of antigen from Leishmania in the skin dermis resulting in a delayed type hypersensitivity (DTH) immune reaction associated with a Th1 immune response and protection against visceral leishmaniasis. Reintroduction of novel approaches for leishmanization and the leishmanin skin test can play a major role in eliminating leishmaniasis.
Collapse
Affiliation(s)
- Thalia Pacheco-Fernandez
- Departments of Pathology and Microbiology, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Greta Volpedo
- Departments of Pathology and Microbiology, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Sreenivas Gannavaram
- Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research (CBER), Food and Drug Administration (FDA), Silver Spring, MD, United States
| | - Parna Bhattacharya
- Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research (CBER), Food and Drug Administration (FDA), Silver Spring, MD, United States
| | - Ranadhir Dey
- Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research (CBER), Food and Drug Administration (FDA), Silver Spring, MD, United States
| | - Abhay Satoskar
- Departments of Pathology and Microbiology, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Greg Matlashewski
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Hira L Nakhasi
- Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research (CBER), Food and Drug Administration (FDA), Silver Spring, MD, United States
| |
Collapse
|
34
|
Soto M, Ramírez L, Solana JC, Cook ECL, Hernández-García E, Charro-Zanca S, Redondo-Urzainqui A, Reguera RM, Balaña-Fouce R, Iborra S. Resistance to Experimental Visceral Leishmaniasis in Mice Infected With Leishmania infantum Requires Batf3. Front Immunol 2020; 11:590934. [PMID: 33362772 PMCID: PMC7758202 DOI: 10.3389/fimmu.2020.590934] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 11/09/2020] [Indexed: 12/20/2022] Open
Abstract
Unveiling the protective immune response to visceral leishmaniasis is critical for a rational design of vaccines aimed at reducing the impact caused by this fatal, if left untreated, vector-borne disease. In this study we sought to determine the role of the basic leucine zipper transcription factor ATF-like 3 (Batf3) in the evolution of infection with Leishmania infantum, the causative agent of human visceral leishmaniasis in the Mediterranean Basin and Latin America. For that, Batf3-deficient mice in C57BL/6 background were infected with an L. infantum strain expressing the luciferase gene. Bioluminescent imaging, as well as in vitro parasite titration, demonstrated that Batf3-deficient mice were unable to control hepatic parasitosis as opposed to wild-type C57BL/6 mice. The impaired microbicide capacities of L. infantum-infected macrophages from Batf3-deficient mice mainly correlated with a reduction of parasite-specific IFN-γ production. Our results reinforce the implication of Batf3 in the generation of type 1 immunity against infectious diseases.
Collapse
Affiliation(s)
- Manuel Soto
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Departamento de Biología Molecular, Nicolás Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain
| | - Laura Ramírez
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Departamento de Biología Molecular, Nicolás Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain
| | - José Carlos Solana
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Departamento de Biología Molecular, Nicolás Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain
| | - Emma C L Cook
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Elena Hernández-García
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Sara Charro-Zanca
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Ana Redondo-Urzainqui
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Rosa M Reguera
- Departamento de Ciencias Biomédicas, Universidad de León, León, Spain
| | | | - Salvador Iborra
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
35
|
Khouili SC, Cook ECL, Hernández-García E, Martínez-López M, Conde-Garrosa R, Iborra S. SHP-1 Regulates Antigen Cross-Presentation and Is Exploited by Leishmania to Evade Immunity. Cell Rep 2020; 33:108468. [PMID: 33264612 DOI: 10.1016/j.celrep.2020.108468] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 10/15/2020] [Accepted: 11/10/2020] [Indexed: 12/12/2022] Open
Abstract
Intracellular pathogens have evolved strategies to evade detection by cytotoxic CD8+ T lymphocytes (CTLs). Here, we ask whether Leishmania parasites trigger the SHP-1-FcRγ chain inhibitory axis to dampen antigen cross-presentation in dendritic cells expressing the C-type lectin receptor Mincle. We find increased cross-priming of CTLs in Leishmania-infected mice deficient for Mincle or with a selective loss of SHP-1 in CD11c+ cells. The latter also shows improved cross-presentation of cell-associated viral antigens. CTL activation in vitro reveals increased MHC class I-peptide complex expression in Mincle- or SHP-1-deficient CD11c+ cells. Neuraminidase treatment also boosts cross-presentation, suggesting that Leishmania triggers SHP-1-associated sialic-acid-binding receptors. Mechanistically, enhanced antigen processing correlates with reduced endosomal acidification in the absence of SHP-1. Finally, we demonstrate that SHP-1 inhibition improves CD11c+ cell-based vaccination against the parasite. Thus, SHP-1-mediated impairment of cross-presentation can be exploited by pathogens to evade CTLs, and SHP-1 inhibition improves CTL responses during vaccination.
Collapse
Affiliation(s)
- Sofía C Khouili
- Immunobiology Lab, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain
| | - Emma C L Cook
- Department of Immunology, School of Medicine, Universidad Complutense de Madrid, 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Elena Hernández-García
- Department of Immunology, School of Medicine, Universidad Complutense de Madrid, 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - María Martínez-López
- Champalimaud Research, Champalimaud Centre for the Unknown, Av. Brasília, 1400-038 Lisboa, Portugal
| | - Ruth Conde-Garrosa
- Immunobiology Lab, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain
| | - Salvador Iborra
- Department of Immunology, School of Medicine, Universidad Complutense de Madrid, 12 de Octubre Health Research Institute (imas12), Madrid, Spain.
| |
Collapse
|
36
|
Reis NFDC, Dupin TV, Costa CR, Toledo MDS, de Oliveira VC, Popi AF, Torrecilhas AC, Xander P. Leishmania amazonensis Promastigotes or Extracellular Vesicles Modulate B-1 Cell Activation and Differentiation. Front Cell Infect Microbiol 2020; 10:573813. [PMID: 33194814 PMCID: PMC7662559 DOI: 10.3389/fcimb.2020.573813] [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: 06/18/2020] [Accepted: 09/28/2020] [Indexed: 12/15/2022] Open
Abstract
B-1 cells are considered an innate-like B cell population that participates in effective innate and adaptive responses to pathogens. B-1 cells produce immunoglobulins, cytokines, chemokines, migrate to inflammatory sites, and differentiate into mononuclear phagocyte-like cells. Murine B-1 cells phagocytosed Leishmaniain vitro and in vivo and participate in immunity against Leishmania. Our group showed that B-1 cells or their extracellular vesicles (EVs) led to a resistance to experimental infection by L. amazonensis. However, the B-1 cells’ responses to Leishmania or EVs isolated from parasites are still poorly characterized. Studying the activation and differentiation of B-1 cells in vivo can contribute to a better understanding of how these cells participate in immunity to L. amazonensis. Thus, we evaluated the expression of myeloid (M-csfr, G-csfr, Spi-1) and lymphoid (EBF, E2A, IL-7R) lineage commitment factors, Toll-like receptors (TLRs), activation cell surface markers, nitric oxide (NO) and reactive oxygen species (ROS) production in murine peritoneal B-1 cells collected after 24 or 48 h post-infection with Leishmania (Leishmania) amazonensis promastigotes or EVs released by the parasites. Our results demonstrated that L. amazonensis infection did not stimulate the expression of CD40, CD80, CD86, F4/80, and MHC II in B-1 cells, but a significant decrease in the production of NO and ROS was observed. The infection induced a significantly higher arginase expression in B-1 cells, but the stimulation with EVs led to a decrease in this gene expression. TLR-2 and TLR-6 had significantly higher expression in B-1 cells from mice intraperitoneally stimulated with the parasite. The TLR-9 expression was higher in animals infected or stimulated for 48 h with EVs. Interestingly, in B-1 cells the stimulus with L. amazonensis led to a substantial increase in the expression of myeloid restricted transcription factors. Thus, our study suggests that the parasites or EVs differently modulated the activation and differentiation of B-1 cells.
Collapse
Affiliation(s)
- Natasha Ferraz de Campos Reis
- Laboratory of Cellular Immunology and Biochemistry of Fungi and Protozoa, Department of Pharmaceutical Sciences, Federal University of São Paulo, São Paulo, Brazil
| | - Talita Vieira Dupin
- Laboratory of Cellular Immunology and Biochemistry of Fungi and Protozoa, Department of Pharmaceutical Sciences, Federal University of São Paulo, São Paulo, Brazil
| | - Carolina Rizzaro Costa
- Laboratory of Cellular Immunology and Biochemistry of Fungi and Protozoa, Department of Pharmaceutical Sciences, Federal University of São Paulo, São Paulo, Brazil
| | - Maytê Dos Santos Toledo
- Laboratory of Cellular Immunology and Biochemistry of Fungi and Protozoa, Department of Pharmaceutical Sciences, Federal University of São Paulo, São Paulo, Brazil
| | - Vivian Cristina de Oliveira
- Department of Microbiology, Immunology and Parasitology, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Ana Flavia Popi
- Department of Microbiology, Immunology and Parasitology, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Ana Claudia Torrecilhas
- Laboratory of Cellular Immunology and Biochemistry of Fungi and Protozoa, Department of Pharmaceutical Sciences, Federal University of São Paulo, São Paulo, Brazil
| | - Patricia Xander
- Laboratory of Cellular Immunology and Biochemistry of Fungi and Protozoa, Department of Pharmaceutical Sciences, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
37
|
Cytokine saga in visceral leishmaniasis. Cytokine 2020; 147:155322. [PMID: 33127259 DOI: 10.1016/j.cyto.2020.155322] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 09/21/2020] [Accepted: 09/25/2020] [Indexed: 12/12/2022]
Abstract
In humans, infection with Leishmania manifests into a spectrum of diseases. The manifestation of the diseases depend on the resultant evasion of the parasite to immune responses namely by macrophages, which is an exclusive host of Leishmania. The B cells valiantly mount antibody responses, however, to no avail as the Leishmania parasites occupy the intracellular niches of the macrophages and subvert the immune response. Extensive studies have been documented on the role of cell-mediated immunity (CMI) in protection and counter survival strategies of the parasites leading to downregulation of CMI. The present review attempts to discuss the cytokines in progression or resolution of visceral form of leishmaniasis or kala-azar, predominantly affecting the Indian subcontinent. The components/cytokine(s) responsible for the regulation of the critical balance of T helper cells and their subsets have been discussed in the perspective. Therefore, any strategy involving the treatment of visceral leishmania (VL) needs to consider the balance and regulation of T cell function.
Collapse
|
38
|
De Silva P, Saad MA, Thomsen HC, Bano S, Ashraf S, Hasan T. Photodynamic therapy, priming and optical imaging: Potential co-conspirators in treatment design and optimization - a Thomas Dougherty Award for Excellence in PDT paper. J PORPHYR PHTHALOCYA 2020; 24:1320-1360. [PMID: 37425217 PMCID: PMC10327884 DOI: 10.1142/s1088424620300098] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
Photodynamic therapy is a photochemistry-based approach, approved for the treatment of several malignant and non-malignant pathologies. It relies on the use of a non-toxic, light activatable chemical, photosensitizer, which preferentially accumulates in tissues/cells and, upon irradiation with the appropriate wavelength of light, confers cytotoxicity by generation of reactive molecular species. The preferential accumulation however is not universal and, depending on the anatomical site, the ratio of tumor to normal tissue may be reversed in favor of normal tissue. Under such circumstances, control of the volume of light illumination provides a second handle of selectivity. Singlet oxygen is the putative favorite reactive molecular species although other entities such as nitric oxide have been credibly implicated. Typically, most photosensitizers in current clinical use have a finite quantum yield of fluorescence which is exploited for surgery guidance and can also be incorporated for monitoring and treatment design. In addition, the photodynamic process alters the cellular, stromal, and/or vascular microenvironment transiently in a process termed photodynamic priming, making it more receptive to subsequent additional therapies including chemo- and immunotherapy. Thus, photodynamic priming may be considered as an enabling technology for the more commonly used frontline treatments. Recently, there has been an increase in the exploitation of the theranostic potential of photodynamic therapy in different preclinical and clinical settings with the use of new photosensitizer formulations and combinatorial therapeutic options. The emergence of nanomedicine has further added to the repertoire of photodynamic therapy's potential and the convergence and co-evolution of these two exciting tools is expected to push the barriers of smart therapies, where such optical approaches might have a special niche. This review provides a perspective on current status of photodynamic therapy in anti-cancer and anti-microbial therapies and it suggests how evolving technologies combined with photochemically-initiated molecular processes may be exploited to become co-conspirators in optimization of treatment outcomes. We also project, at least for the short term, the direction that this modality may be taking in the near future.
Collapse
Affiliation(s)
- Pushpamali De Silva
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Mohammad A. Saad
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Hanna C. Thomsen
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Shazia Bano
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Shoaib Ashraf
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Division of Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
39
|
Dos Santos JC, Barroso de Figueiredo AM, Teodoro Silva MV, Cirovic B, de Bree LCJ, Damen MSMA, Moorlag SJCFM, Gomes RS, Helsen MM, Oosting M, Keating ST, Schlitzer A, Netea MG, Ribeiro-Dias F, Joosten LAB. β-Glucan-Induced Trained Immunity Protects against Leishmania braziliensis Infection: a Crucial Role for IL-32. Cell Rep 2020; 28:2659-2672.e6. [PMID: 31484076 DOI: 10.1016/j.celrep.2019.08.004] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 06/04/2019] [Accepted: 07/30/2019] [Indexed: 01/11/2023] Open
Abstract
American tegumentary leishmaniasis is a vector-borne parasitic disease caused by Leishmania protozoans. Innate immune cells undergo long-term functional reprogramming in response to infection or Bacillus Calmette-Guérin (BCG) vaccination via a process called trained immunity, conferring non-specific protection from secondary infections. Here, we demonstrate that monocytes trained with the fungal cell wall component β-glucan confer enhanced protection against infections caused by Leishmania braziliensis through the enhanced production of proinflammatory cytokines. Mechanistically, this augmented immunological response is dependent on increased expression of interleukin 32 (IL-32). Studies performed using a humanized IL-32 transgenic mouse highlight the clinical implications of these findings in vivo. This study represents a definitive characterization of the role of IL-32γ in the trained phenotype induced by β-glucan or BCG, the results of which improve our understanding of the molecular mechanisms governing trained immunity and Leishmania infection control.
Collapse
Affiliation(s)
- Jéssica Cristina Dos Santos
- Radboud Institute for Molecular Sciences (RILMS), Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands; Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | | | | | - Branko Cirovic
- Myeloid Cell Biology, Life and Medical Sciences Institute, University of Bonn, 53115 Bonn, Germany
| | - L Charlotte J de Bree
- Radboud Institute for Molecular Sciences (RILMS), Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands; Research Center for Vitamins and Vaccines, Bandim Health Project, Statens Serum Institut, Copenhagen, Denmark; Odense Patient Data Explorative Network, University of Southern Denmark and Odense University Hospital, Odense, Denmark
| | - Michelle S M A Damen
- Radboud Institute for Molecular Sciences (RILMS), Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Simone J C F M Moorlag
- Radboud Institute for Molecular Sciences (RILMS), Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Rodrigo S Gomes
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Monique M Helsen
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Marije Oosting
- Radboud Institute for Molecular Sciences (RILMS), Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Samuel T Keating
- Radboud Institute for Molecular Sciences (RILMS), Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - A Schlitzer
- Myeloid Cell Biology, Life and Medical Sciences Institute, University of Bonn, 53115 Bonn, Germany; Single Cell Genomics and Epigenomics Unit at the German Center for Neurodegenerative Diseases and the University of Bonn, 53175 Bonn, Germany
| | - Mihai G Netea
- Radboud Institute for Molecular Sciences (RILMS), Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands; Department for Genomics and Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Germany
| | - Fátima Ribeiro-Dias
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás, Brazil.
| | - Leo A B Joosten
- Radboud Institute for Molecular Sciences (RILMS), Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands; Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás, Brazil.
| |
Collapse
|
40
|
Saunders EC, McConville MJ. Immunometabolism of Leishmania granulomas. Immunol Cell Biol 2020; 98:832-844. [PMID: 32780446 DOI: 10.1111/imcb.12394] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/06/2020] [Accepted: 08/06/2020] [Indexed: 12/19/2022]
Abstract
Leishmania are parasitic protists that cause a spectrum of diseases in humans characterized by the formation of granulomatous lesions in the skin or other tissues, such as liver and spleen. The extent to which Leishmania granulomas constrain or promote parasite growth is critically dependent on the host T-helper type 1/T-helper type 2 immune response and the localized functional polarization of infected and noninfected macrophages toward a classically (M1) or alternatively (M2) activated phenotype. Recent studies have shown that metabolic reprograming of M1 and M2 macrophages underpins the capacity of these cells to act as permissive or nonpermissive host reservoirs, respectively. In this review, we highlight the metabolic requirements of Leishmania amastigotes and the evidence that these parasites induce and/or exploit metabolic reprogramming of macrophage metabolism. We also focus on recent studies highlighting the role of key macrophage metabolic signaling pathways, such as mechanistic target of rapamycin, adenosine monophosphate-activated protein kinase and peroxisome proliferator receptor gamma in regulating the pathological progression of Leishmania granulomas. These studies highlight the intimate connectivity between Leishmania and host cell metabolism, the need to investigate these interactions in vivo and the potential to exploit host cell metabolic signaling pathways in developing new host-directed therapies.
Collapse
Affiliation(s)
- Eleanor C Saunders
- Department of Biochemistry and Molecular Biology, Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Malcolm J McConville
- Department of Biochemistry and Molecular Biology, Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Parkville, VIC, 3010, Australia
| |
Collapse
|
41
|
Santos MF, Alexandre-Pires G, Pereira MA, Gomes L, Rodrigues AV, Basso A, Reisinho A, Meireles J, Santos-Gomes GM, Pereira da Fonseca I. Immunophenotyping of Peripheral Blood, Lymph Node, and Bone Marrow T Lymphocytes During Canine Leishmaniosis and the Impact of Antileishmanial Chemotherapy. Front Vet Sci 2020; 7:375. [PMID: 32760744 PMCID: PMC7373748 DOI: 10.3389/fvets.2020.00375] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 05/28/2020] [Indexed: 11/30/2022] Open
Abstract
Dogs are a major reservoir of Leishmania infantum, etiological agent of canine leishmaniosis (CanL) a zoonotic visceral disease of worldwide concern. Therapeutic protocols based on antileishmanial drugs are commonly used to treat sick dogs and improve their clinical condition. To better understand the impact of Leishmania infection and antileishmanial drugs on the dog's immune response, this study investigates the profile of CD4+ and CD8+ T cell subsets in peripheral blood, lymph node, and bone marrow of sick dogs and after two different CanL treatments. Two CanL groups of six dogs each were treated with either miltefosine or meglumine antimoniate combined with allopurinol. Another group of 10 clinically healthy dogs was used as control. Upon diagnosis and during the following 3 months of treatment, peripheral blood, popliteal lymph node, and bone marrow mononuclear cells were collected, labeled for surface markers CD45, CD3, CD4, CD8, CD25, and intracellular nuclear factor FoxP3, and T lymphocyte subpopulations were immunophenotyped by flow cytometry. CanL dogs presented an overall increased frequency of CD8+ and CD4+CD8+ double-positive T cells in all tissues and a decreased frequency of CD4+ T cells in the blood. Furthermore, there was a higher frequency of CD8+ T cells expressing CD25+FoxP3+ in the blood and bone marrow. During treatment, these subsets recovered to levels similar to those of healthy dogs. Nevertheless, antileishmanial therapy caused an increase of CD4+CD25+FoxP3+ T cells in all tissues, associated with the decrease of CD8+CD25−FoxP3− T cell percentages. These findings may support previous studies that indicate that L. infantum manipulates the dog's immune system to avoid the development of a protective response, ensuring the parasite's survival and the conditions that allow the completion of Leishmania life cycle. Both treatments used appear to have an effect on the dog's immune response, proving to be effective in promoting the normalization of T cell subsets.
Collapse
Affiliation(s)
- Marcos Ferreira Santos
- CIISA-Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Lisbon, Portugal
| | - Graça Alexandre-Pires
- CIISA-Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Lisbon, Portugal
| | - Maria A Pereira
- GHTM-Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova De Lisbon (UNL), Lisbon, Portugal
| | - Lídia Gomes
- CIISA-Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Lisbon, Portugal
| | - Armanda V Rodrigues
- GHTM-Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova De Lisbon (UNL), Lisbon, Portugal
| | - Alexandra Basso
- CIISA-Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Lisbon, Portugal
| | - Ana Reisinho
- CIISA-Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Lisbon, Portugal
| | - José Meireles
- CIISA-Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Lisbon, Portugal
| | - Gabriela M Santos-Gomes
- GHTM-Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova De Lisbon (UNL), Lisbon, Portugal
| | - Isabel Pereira da Fonseca
- CIISA-Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
42
|
Lecoeur H, Rosazza T, Kokou K, Varet H, Coppée JY, Lari A, Commère PH, Weil R, Meng G, Milon G, Späth GF, Prina E. Leishmania amazonensis Subverts the Transcription Factor Landscape in Dendritic Cells to Avoid Inflammasome Activation and Stall Maturation. Front Immunol 2020; 11:1098. [PMID: 32582184 PMCID: PMC7295916 DOI: 10.3389/fimmu.2020.01098] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 05/06/2020] [Indexed: 12/18/2022] Open
Abstract
Leishmania parasites are the causative agents of human leishmaniases. They infect professional phagocytes of their mammalian hosts, including dendritic cells (DCs) that are essential for the initiation of adaptive immune responses. These immune functions strictly depend on the DC's capacity to differentiate from immature, antigen-capturing cells to mature, antigen-presenting cells—a process accompanied by profound changes in cellular phenotype and expression profile. Only little is known on how intracellular Leishmania affects this important process and DC transcriptional regulation. Here, we investigate these important open questions analyzing phenotypic, cytokine profile and transcriptomic changes in murine, immature bone marrow-derived DCs (iBMDCs) infected with antibody-opsonized and non-opsonized Leishmania amazonensis (L.am) amastigotes. DCs infected by non-opsonized amastigotes remained phenotypically immature whereas those infected by opsonized parasites displayed a semi-mature phenotype. The low frequency of infected DCs in culture led us to use DsRed2-transgenic parasites allowing for the enrichment of infected BMDCs by FACS. Sorted infected DCs were then subjected to transcriptomic analyses using Affymetrix GeneChip technology. Independent of parasite opsonization, Leishmania infection induced expression of genes related to key DC processes involved in MHC Class I-restricted antigen presentation and alternative NF-κB activation. DCs infected by non-opsonized parasites maintained an immature phenotype and showed a small but significant down-regulation of gene expression related to pro-inflammatory TLR signaling, the canonical NF-kB pathway and the NLRP3 inflammasome. This transcriptomic profile was further enhanced in DCs infected with opsonized parasites that displayed a semi-mature phenotype despite absence of inflammasome activation. This paradoxical DC phenotype represents a Leishmania-specific signature, which to our knowledge has not been observed with other opsonized infectious agents. In conclusion, systems-analyses of our transcriptomics data uncovered important and previously unappreciated changes in the DC transcription factor landscape, thus revealing a novel Leishmania immune subversion strategy directly acting on transcriptional control of gene expression. Our data raise important questions on the dynamic and reciprocal interplay between trans-acting and epigenetic regulators in establishing permissive conditions for intracellular Leishmania infection and polarization of the immune response.
Collapse
Affiliation(s)
- Hervé Lecoeur
- Institut Pasteur, INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Département des Parasites et Insectes Vecteurs, Paris, France.,Pasteur Institute of Shanghai, Innate Immunity Unit, Key Laboratory of Molecular Virology and Immunology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,Pasteur International Unit "Inflammation and Leishmania Infection", Paris, France
| | - Thibault Rosazza
- Institut Pasteur, INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Département des Parasites et Insectes Vecteurs, Paris, France.,Pasteur International Unit "Inflammation and Leishmania Infection", Paris, France
| | - Kossiwa Kokou
- Institut Pasteur, INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Département des Parasites et Insectes Vecteurs, Paris, France.,Pasteur Institute of Shanghai, Innate Immunity Unit, Key Laboratory of Molecular Virology and Immunology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,Pasteur International Unit "Inflammation and Leishmania Infection", Paris, France
| | - Hugo Varet
- Hub de Bioinformatique et Biostatistique - Département Biologie Computationnelle, Institut Pasteur, USR 3756 CNRS, Paris, France
| | - Jean-Yves Coppée
- Institut Pasteur - Transcriptome and Epigenome Platform - Biomics Pole - C2RT, Paris, France
| | - Arezou Lari
- Systems Biomedicine Unit, Institut Pasteur of Iran, Teheran, Iran
| | | | - Robert Weil
- Sorbonne Universités, Institut National de la Santé et de la Recherche Médicale (Inserm, UMR1135), Centre National de la Recherche Scientifique (CNRS, ERL8255), Centre d'Immunologie et des Maladies Infectieuses CIMI, Paris, France
| | - Guangxun Meng
- Pasteur Institute of Shanghai, Innate Immunity Unit, Key Laboratory of Molecular Virology and Immunology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,Pasteur International Unit "Inflammation and Leishmania Infection", Paris, France
| | - Genevieve Milon
- Institut Pasteur, Laboratoire Immunophysiologie et Parasitisme, Département des Parasites et Insectes Vecteurs, Paris, France
| | - Gerald F Späth
- Institut Pasteur, INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Département des Parasites et Insectes Vecteurs, Paris, France.,Pasteur International Unit "Inflammation and Leishmania Infection", Paris, France
| | - Eric Prina
- Institut Pasteur, INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Département des Parasites et Insectes Vecteurs, Paris, France.,Pasteur International Unit "Inflammation and Leishmania Infection", Paris, France
| |
Collapse
|
43
|
Taslimi Y, Agbajogu C, Brynjolfsson SF, Masoudzadeh N, Mashayekhi V, Gharibzadeh S, Östensson M, Nakka SS, Mizbani A, Rafati S, Harandi AM. Profiling inflammatory response in lesions of cutaneous leishmaniasis patients using a non-invasive sampling method combined with a high-throughput protein detection assay. Cytokine 2020; 130:155056. [PMID: 32199248 DOI: 10.1016/j.cyto.2020.155056] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/23/2020] [Accepted: 02/27/2020] [Indexed: 12/25/2022]
Abstract
BACKGROUND Cutaneous leishmaniasis (CL) is an infection caused by Leishmania (L.) protozoa transmitted through the bite of infected sand fly. Previously, invasive sampling of blood and skin along with low throughput methods were used for determination of inflammatory response in CL patients. AIMS/METHODOLOGY We established a novel approach based on a non-invasive adhesive tape-disc sampling combined with a powerful multiplexing technique called proximity extension assay for profiling 92 inflammatory cytokines, chemokines and surface molecules in the lesions of CL patients infected with L. tropica. Sample collection was done non-invasively by using adhesive tape-discs from lesion and normal skin of 33 L. tropica positive patients. RESULTS Out of 92 inflammatory proteins, the level of 34 proteins was significantly increased in the lesions of CL patients compared to their normal skin. This includes the chemokines CCL2, CCL3, CCL4, CXCL1, CXCL5, CXCL9, CXCL10 and CXCL11, together with the interleukins IL-6, IL-8, IL-18, LIF and OSM. The remaining significantly changed inflammatory proteins include 7 surface molecules and receptors: CD5, CD40, CDCP1, 4E-BP1, TNFRSF9, IL-18R1 and OPG as well as 16 other cytokines and proteins: MMP-1, CSF-1, VEGFA, uPA, EN-RAGE, LAP TGF-β1, HGF, MMP-10, CASP-8, TNFSF14, STAMPB, ADA, TRAIL and ST1A1. Further, 13 proteins showed an increasing trend, albeit not statistically significant, in the CL lesions, including TGF-α, CCL23, MCP-2, IL-12B, CXCL6, IL-24, FGF-19, TNFβ, CD6, TRANCE, IL10, SIR2 and CCL20. CONCLUSION We herein report a novel approach based on a non-invasive sampling method combined with the high-throughput protein assay for profiling inflammatory proteins in CL lesions. Using this approach, we could profile inflammatory proteins in the lesions from CL patients. This new non-invasive approach may have implications for studying skin inflammatory mediators in CL and other skin disorders.
Collapse
Affiliation(s)
- Yasaman Taslimi
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | - Christopher Agbajogu
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | | | - Nasrin Masoudzadeh
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | - Vahid Mashayekhi
- Cutaneous Leishmaniasis Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Safoora Gharibzadeh
- Department of Epidemiology and Biostatistics, Research Centre for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran
| | - Malin Östensson
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Sravya Sowdamini Nakka
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | | | - Sima Rafati
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran.
| | - Ali M Harandi
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden; Vaccine Evaluation Center, BC Children's Hospital Research Institute, The University of British Columbia, Canada.
| |
Collapse
|
44
|
Hurdayal R, Nieuwenhuizen NE, Khutlang R, Brombacher F. Inflammatory Dendritic Cells, Regulated by IL-4 Receptor Alpha Signaling, Control Replication, and Dissemination of Leishmania major in Mice. Front Cell Infect Microbiol 2020; 9:479. [PMID: 32039054 PMCID: PMC6992597 DOI: 10.3389/fcimb.2019.00479] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 12/27/2019] [Indexed: 11/16/2022] Open
Abstract
Leishmaniasis is a vector-borne disease caused by Leishmania parasites. Macrophages are considered the primary parasite host cell, but dendritic cells (DCs) play a critical role in initiating adaptive immunity and controlling Leishmania infection. Accordingly, our previous study in CD11ccreIL-4Rα−/lox mice, which have impaired IL-4 receptor alpha (IL-4Rα) expression on CD11c+ cells including DCs, confirmed a protective role for IL-4/IL-13-responsive DCs in replication and dissemination of parasites during cutaneous leishmaniasis. However, it was unclear which DC subset/s was executing this function. To investigate this, we infected CD11ccreIL-4Rα−/lox and control mice with L. major GFP+ parasites and identified subsets of infected DCs by flow cytometry. Three days after infection, CD11b+ DCs and CD103+ DCs were the main infected DC subsets in the footpad and draining lymph node, respectively and by 4 weeks post-infection, Ly6C+ and Ly6C− CD11b+ DCs were the main infected DC populations in both the lymph nodes and footpads. Interestingly, Ly6C+CD11b+ inflammatory monocyte-derived DCs but not Ly6C−CD11b+ DCs hosted parasites in the spleen. Importantly, intracellular parasitism was significantly higher in IL-4Rα-deficient DCs. In terms of DC effector function, we found no change in the expression of pattern-recognition receptors (TLR4 and TLR9) nor in expression of the co-stimulatory marker, CD80, but MHCII expression was lower in CD11ccreIL-4Rα−/lox mice at later time-points compared to the controls. Interestingly, in CD11ccreIL-4Rα−/lox mice, which have reduced Th1 responses, CD11b+ DCs had impaired iNOS production, suggesting that DC IL-4Rα expression and NO production is important for controlling parasite numbers and preventing dissemination. Expression of the alternative activation marker arginase was unchanged in CD11b+ DCs in CD11creIL-4Rα−/lox mice compared to littermate controls, but RELM-α was upregulated, suggesting IL-4Rα-independent alternative activation. In summary, L. major parasites may use Ly6C+CD11b+ inflammatory DCs derived from monocytes recruited to infection as “Trojan horses” to migrate to secondary lymphoid organs and peripheral sites, and DC IL-4Rα expression is important for controlling infection.
Collapse
Affiliation(s)
- Ramona Hurdayal
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, South Africa.,International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Cape Town, South Africa.,Division of Immunology, Department of Pathology, Faculty of Health Sciences, South African Medical Research Council on Immunology of Infectious Diseases, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Faculty of Health Sciences, Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Natalie Eva Nieuwenhuizen
- International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Cape Town, South Africa.,Division of Immunology, Department of Pathology, Faculty of Health Sciences, South African Medical Research Council on Immunology of Infectious Diseases, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Rethabile Khutlang
- Identity Authentication Research Group, Defence and Security, Council for Scientific and Industrial Research, Pretoria, South Africa
| | - Frank Brombacher
- International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Cape Town, South Africa.,Division of Immunology, Department of Pathology, Faculty of Health Sciences, South African Medical Research Council on Immunology of Infectious Diseases, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Faculty of Health Sciences, Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
45
|
Handschuh J, Amore J, Müller AJ. From the Cradle to the Grave of an Infection: Host-Pathogen Interaction Visualized by Intravital Microscopy. Cytometry A 2019; 97:458-470. [PMID: 31777152 DOI: 10.1002/cyto.a.23938] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/12/2019] [Accepted: 11/06/2019] [Indexed: 12/11/2022]
Abstract
During infections, interactions between host immune cells and the pathogen occur in distinct anatomical locations and along defined time scales. This can best be assessed in the physiological context of an infection in the living tissue. Consequently, intravital imaging has enabled us to dissect the critical phases and events throughout an infection in real time in living tissues. Specifically, advances in visualizing specific cell types and individual pathogens permitted tracking the early events of tissue invasion of the pathogen, cellular interactions involved in the induction of the immune response as well the events implicated in clearance of the infection. In this respect, two vantage points have evolved since the initial employment of this technique in the field of infection biology. On the one hand, strategies acquired by the pathogen to establish within the host and circumvent or evade the immune defenses have been elucidated. On the other hand, analyzing infections from the immune system's perspective has led to insights into the dynamic cellular interactions that are involved in the initial recognition of the pathogen, immune induction as well as effector function delivery and immunopathology. Furthermore, an increasing interest in probing functional parameters in vivo has emerged, such as the analysis of pathogen reactivity to stress conditions imposed by the host organism in order to mediate clearance upon pathogen encounter. Here, we give an overview on recent intravital microscopy findings of host-pathogen interactions along the course of an infection, from both the immune system's and pathogen's perspectives. We also discuss recent developments and future perspectives in extracting intravital information beyond the localization of pathogens and their interaction with immune cells. Such reporter systems on the pathogen's physiological state and immune cell functions may prove useful in dissecting the functional dynamics of host-pathogen interactions. © 2019 The Authors. Cytometry Part A published by Wiley Periodicals, Inc. on behalf of International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Juliane Handschuh
- Institute of Molecular and Clinical Immunology, Health Campus Immunology Infectiology and Inflammation (GC-I3), Otto-von-Guericke-University, 39120, Magdeburg, Germany
| | - Jonas Amore
- Institute of Molecular and Clinical Immunology, Health Campus Immunology Infectiology and Inflammation (GC-I3), Otto-von-Guericke-University, 39120, Magdeburg, Germany
| | - Andreas J Müller
- Institute of Molecular and Clinical Immunology, Health Campus Immunology Infectiology and Inflammation (GC-I3), Otto-von-Guericke-University, 39120, Magdeburg, Germany.,Intravital Microscopy of Infection and Immunity, Helmholtz Centre for Infection Research, 38124, Braunschweig, Germany
| |
Collapse
|
46
|
da Silva Vieira T, Arango Duque G, Ory K, Gontijo CM, Soares RP, Descoteaux A. Leishmania braziliensis: Strain-Specific Modulation of Phagosome Maturation. Front Cell Infect Microbiol 2019; 9:319. [PMID: 31555609 PMCID: PMC6743224 DOI: 10.3389/fcimb.2019.00319] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 08/26/2019] [Indexed: 12/17/2022] Open
Abstract
Leishmania (Viannia) braziliensis is responsible for the largest number of American tegumentary leishmaniasis (ATL) in Brazil. ATL can present several clinical forms including typical (TL) and atypical (AL) cutaneous and mucocutaneous (ML) lesions. To identify parasite and host factors potentially associated with these diverse clinical manifestations, we first surveyed the expression of two virulence-associated glycoconjugates, lipophosphoglycan (LPG) and the metalloprotease GP63 by a panel of promastigotes of Leishmania braziliensis (L. braziliensis) strains isolated from patients with different clinical manifestations of ATL and from the sand fly vector. We observed a diversity of expression patterns for both LPG and GP63, which may be related to strain-specific polymorphisms. Interestingly, we noted that GP63 activity varies from strain to strain, including the ability to cleave host cell molecules. We next evaluated the ability of promastigotes from these L. braziliensis strains to modulate phagolysosome biogenesis in bone marrow-derived macrophages (BMM), by assessing phagosomal recruitment of the lysosome-associated membrane protein 1 (LAMP-1) and intraphagosomal acidification. Whereas, three out of six L. braziliensis strains impaired the phagosomal recruitment of LAMP-1, only the ML strain inhibited phagosome acidification to the same extent as the L. donovani strain that was used as a positive control. While decreased phagosomal recruitment of LAMP-1 correlated with higher LPG levels, decreased phagosomal acidification correlated with higher GP63 levels. Finally, we observed that the ability to infect and replicate within host cells did not fully correlate with the inhibition of phagosome maturation. Collectively, our results revealed a diversity of strain-specific phenotypes among L. braziliensis isolates, consistent with the high genetic diversity within Leishmania populations.
Collapse
Affiliation(s)
- Tamara da Silva Vieira
- Fundação Oswaldo Cruz - FIOCRUZ, Centro de Pesquisas René Rachou, Belo Horizonte, Brazil.,INRS - Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Laval, QC, Canada
| | - Guillermo Arango Duque
- INRS - Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Laval, QC, Canada
| | - Kévin Ory
- INRS - Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Laval, QC, Canada.,Université de Rennes 1, CHU Rennes, INSERM, Rennes, France
| | - Celia Maria Gontijo
- Fundação Oswaldo Cruz - FIOCRUZ, Centro de Pesquisas René Rachou, Belo Horizonte, Brazil
| | - Rodrigo Pedro Soares
- Fundação Oswaldo Cruz - FIOCRUZ, Centro de Pesquisas René Rachou, Belo Horizonte, Brazil
| | - Albert Descoteaux
- INRS - Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Laval, QC, Canada
| |
Collapse
|
47
|
Oualha R, Barhoumi M, Marzouki S, Harigua-Souiai E, Ben Ahmed M, Guizani I. Infection of Human Neutrophils With Leishmania infantum or Leishmania major Strains Triggers Activation and Differential Cytokines Release. Front Cell Infect Microbiol 2019; 9:153. [PMID: 31134162 PMCID: PMC6524560 DOI: 10.3389/fcimb.2019.00153] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 04/24/2019] [Indexed: 12/27/2022] Open
Abstract
Leishmaniases are neglected diseases, caused by intracellular protozoan parasites of the Leishmania (L.) genus. Although the principal host cells of the parasites are macrophages, neutrophils are the first cells rapidly recruited to the site of parasites inoculation, where they play an important role in the early recognition and elimination of the parasites. The nature of early interactions between neutrophils and Leishmania could influence the outcome of infection. Herein we aimed to evaluate whether different Leishmania strains, responsible for distinct clinical manifestations, could influence ex vivo functional activity of neutrophils. Human polymorphonuclear leukocytes were isolated from 14 healthy volunteers and the ex vivo infection of these cells was done with two L. infantum and one L. major strains. Infection parameters were determined and neutrophils activation was assessed by oxidative burst, degranulation, DNA release and apoptosis; cytokine production was measured by a multiplex flow cytometry analysis. Intracellular amastigotes were rescued to determine Leishmania strains survival. The results showed that L. infantum and L. major promastigotes similarly infected the neutrophils. Oxidative burst, neutrophil elastase, myeloperoxidase activity and apoptosis were significantly increased in infected neutrophils but with no differences between strains. The L. infantum-infected neutrophils induced more DNA release than those infected by L. major. Furthermore, Leishmania strains induced high amounts of IL-8 and stimulated the production of IL-1β, TNF-α, and TGF-β by human neutrophils. We observed that only one strain promoted IL-6 release by these neutrophils. The production of TNF-α was also differently induced by the parasites strains. All these results demonstrate that L. infantum and L. major strains were able to induce globally a similar ex vivo activation and apoptosis of neutrophils; however, they differentially triggered cytokines release from these cells. In addition, rescue of intracellular parasites indicated different survival rates further emphasizing on the influence of parasite strains within a species on the fate of infection.
Collapse
Affiliation(s)
- Rafeh Oualha
- Laboratory of Molecular Epidemiology and Experimental Pathology - LR16IPT04, Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis, Tunisia.,Faculté des Sciences de Bizerte, Université de Carthage, Tunis, Tunisia
| | - Mourad Barhoumi
- Laboratory of Molecular Epidemiology and Experimental Pathology - LR16IPT04, Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Soumaya Marzouki
- Laboratory of Transmission, Control and Immunobiology of Infections - LR16IPT02, Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Emna Harigua-Souiai
- Laboratory of Molecular Epidemiology and Experimental Pathology - LR16IPT04, Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Melika Ben Ahmed
- Laboratory of Transmission, Control and Immunobiology of Infections - LR16IPT02, Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Ikram Guizani
- Laboratory of Molecular Epidemiology and Experimental Pathology - LR16IPT04, Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis, Tunisia
| |
Collapse
|
48
|
Afrin F, Khan I, Hemeg HA. Leishmania-Host Interactions-An Epigenetic Paradigm. Front Immunol 2019; 10:492. [PMID: 30967861 PMCID: PMC6438953 DOI: 10.3389/fimmu.2019.00492] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 02/25/2019] [Indexed: 12/17/2022] Open
Abstract
Leishmaniasis is one of the major neglected tropical diseases, for which no vaccines exist. Chemotherapy is hampered by limited efficacy coupled with development of resistance and other side effects. Leishmania parasites elude the host defensive mechanisms by modulating their surface proteins as well as dampening the host's immune responses. The parasites use the conventional RNA polymerases peculiarly under different environmental cues or pressures such as the host's milieu or the drugs. The mechanisms that restructure post-translational modifications are poorly understood but altered epigenetic histone modifications are believed to be instrumental in influencing the chromatin remodeling in the parasite. Interestingly, the parasite also modulates gene expression of the hosts, thereby hijacking or dampening the host immune response. Epigenetic factor such as DNA methylation of cytosine residues has been incriminated in silencing of macrophage-specific genes responsible for defense against these parasites. Although there is dearth of information regarding the epigenetic alterations-mediated pathogenesis in these parasites and the host, the unique epigenetic marks may represent targets for potential anti-leishmanial drug candidates. This review circumscribes the epigenetic changes during Leishmania infection, and the epigenetic modifications they enforce upon the host cells to ensure a safe haven. The non-coding micro RNAs as post-transcriptional regulators and correlates of wound healing and toll-like receptor signaling, as well as prognostic biomarkers of therapeutic failure and healing time are also explored. Finally, we highlight the recent advances on how the epigenetic perturbations may impact leishmaniasis vaccine development as biomarkers of safety and immunogenicity.
Collapse
Affiliation(s)
- Farhat Afrin
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Taibah University, Madina, Saudi Arabia
| | - Inbesat Khan
- Rajiv Gandhi Technical University, Bhopal, India
| | - Hassan A Hemeg
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Taibah University, Madina, Saudi Arabia
| |
Collapse
|
49
|
Mollinedo F. Neutrophil Degranulation, Plasticity, and Cancer Metastasis. Trends Immunol 2019; 40:228-242. [PMID: 30777721 DOI: 10.1016/j.it.2019.01.006] [Citation(s) in RCA: 224] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 01/13/2019] [Accepted: 01/14/2019] [Indexed: 02/08/2023]
Abstract
Neutrophils are the first responders to inflammation and infection. Recently, an elevated neutrophil-to-lymphocyte ratio has generally become a prognostic indicator of poor overall survival in cancer. Accordingly, heterogeneous ill-defined neutrophil-like populations have been increasingly recognized as important players in cancer development. In addition, neutrophil granule proteins released upon cell activation have been associated with tumor progression; this differential granule mobilization may allow neutrophils - and possibly associated cancer cells - to leave the bloodstream and enter inflamed/infected tissues. This review discusses and proposes how granule mobilization may facilitate neutrophil-mediated transport of cancer cells into different tissues as well as leading to different cellular phenotypes that underlie remarkable neutrophil plasticity. This concept might inform novel neutrophil-centered approaches to putative cancer therapies.
Collapse
Affiliation(s)
- Faustino Mollinedo
- Laboratory of Cell Death and Cancer Therapy, Department of Molecular Biomedicine, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), Calle Ramiro de Maeztu 9, E-28040 Madrid, Spain.
| |
Collapse
|
50
|
Wong J, Choi SYC, Liu R, Xu E, Killam J, Gout PW, Wang Y. Potential Therapies for Infectious Diseases Based on Targeting Immune Evasion Mechanisms That Pathogens Have in Common With Cancer Cells. Front Cell Infect Microbiol 2019; 9:25. [PMID: 30809511 PMCID: PMC6379255 DOI: 10.3389/fcimb.2019.00025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 01/24/2019] [Indexed: 12/18/2022] Open
Abstract
Many global infectious diseases are not well-controlled, underlining a critical need for new, more effective therapies. Pathogens and pathogen-infected host cells, like cancer cells, evade immune surveillance via immune evasion mechanisms. The present study indicates that pathogenic bacteria, endoparasites, and virus-infected host cells can have immune evasion mechanisms in common with cancers. These include entry into dormancy and metabolic reprogramming to aerobic glycolysis leading to excessive secretion of lactic acid and immobilization of local host immunity. The latter evasion tactic provides a therapeutic target for cancer, as shown by our recent finding that patient-derived cancer xenografts can be growth-arrested, without major host toxicity, by inhibiting their lactic acid secretion (as mediated by the MCT4 transporter)-with evidence of host immunity restoration. Accordingly, the multiplication of bacteria, endoparasites, and viruses that primarily depend on metabolic reprogramming to aerobic glycolysis for survival may be arrested using cancer treatment strategies that inhibit their lactic acid secretion. Immune evasion mechanisms shared by pathogens and cancer cells likely represent fundamental, evolutionarily-conserved mechanisms that may be particularly critical to their welfare. As such, their targeting may lead to novel therapies for infectious diseases.
Collapse
Affiliation(s)
- Jodi Wong
- Department of Experimental Therapeutics, BC Cancer Research Centre, Vancouver, BC, Canada
| | - Stephen Yiu Chuen Choi
- Department of Experimental Therapeutics, BC Cancer Research Centre, Vancouver, BC, Canada.,Vancouver Prostate Centre, Vancouver, BC, Canada.,Department of Urologic Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Rongrong Liu
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Eddie Xu
- Department of Experimental Therapeutics, BC Cancer Research Centre, Vancouver, BC, Canada.,Vancouver Prostate Centre, Vancouver, BC, Canada.,Department of Urologic Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - James Killam
- Vancouver Prostate Centre, Vancouver, BC, Canada
| | - Peter W Gout
- Department of Experimental Therapeutics, BC Cancer Research Centre, Vancouver, BC, Canada
| | - Yuzhuo Wang
- Department of Experimental Therapeutics, BC Cancer Research Centre, Vancouver, BC, Canada.,Vancouver Prostate Centre, Vancouver, BC, Canada.,Department of Urologic Sciences, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|