1
|
Sebastian P, Namdeo M, Devender M, Anand A, Kumar K, Veronica J, Maurya R. Polyamine-Enriched Exosomes from Leishmania donovani Drive Host Macrophage Polarization via Immunometabolism Reprogramming. ACS Infect Dis 2024; 10:4384-4399. [PMID: 39560603 DOI: 10.1021/acsinfecdis.4c00738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2024]
Abstract
Leishmania donovani (Ld) promastigotes secrete exosomes that are crucial in host-pathogen interactions and intercellular communication by carrying parasite-specific molecules. Although the composition of cargos in Leishmania exosomes is known, the effects of the unique metabolic repertoire on immunometabolism rewiring of macrophage polarization are poorly understood. Interestingly, we found the enrichment of polyamines (PAs) such as spermidine and putrescine in the Ld-exosomes. Herein, we investigate the critical polycationic molecules and their crucial role in parasite survival. Our study shows that PA inhibition or depletion significantly impairs parasite growth and fitness, particularly in drug-resistant strains. Furthermore, we aimed to elucidate the impact of PAs-enriched Ld-exosomes on host macrophages. The data demonstrated that macrophages efficiently internalized these exosomes, leading to heightened phagocytic activity and infectivity. In addition, internalized Ld-exosomes induced M2 macrophage polarization characterized by elevated Arginase-1 expression and activity. The increased expression of the solute carrier gene (SLC3A2) and elevated intracellular spermidine levels suggest that Ld-exosomes contribute to the host PAs pool and create an anti-inflammatory milieu. These findings highlight the essential role of PAs-enriched Ld-exosomes in parasite survival and establishing a pro-parasitic environment in the host macrophage.
Collapse
Affiliation(s)
- Prince Sebastian
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India
| | - Madhulika Namdeo
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India
| | - Moodu Devender
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India
| | - Anjali Anand
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India
| | - Krishan Kumar
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India
| | - Jalaja Veronica
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India
| | - Radheshyam Maurya
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India
| |
Collapse
|
2
|
Ghosh R, Bishayi B. Neutralization of TLR2 in combination with either TNF-α or IL-1β antibody reduces the severity of septic arthritis through STAT3/mTOR signalling in lymphocytes. Cell Immunol 2024; 405-406:104878. [PMID: 39312873 DOI: 10.1016/j.cellimm.2024.104878] [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: 07/06/2024] [Revised: 08/23/2024] [Accepted: 09/13/2024] [Indexed: 09/25/2024]
Abstract
Staphylococcus aureus induced Septic arthritis is considered a medical concern. S.aureus binds TLR2 to induce an array of inflammatory responses. Generation of pro-inflammatory cytokines induces T cell responses and control Th17/Treg cell balance. Regulation of T cell-mediated immunity in response to inflammation is significantly influenced by mTOR. Presence of elevated TNF-α, IL-1β decreases Treg cell activity through STAT3/mTOR, promoting proliferation of T cells towards Th17 cells. Therefore, we postulated, neutralizing TLR2 with either TNF-α or IL-1β in combination could be useful in modifying Th17/Treg cell ratio in order to treat septic arthritis by suppressing expression of mTOR/STAT3. To date, no studies have reported effects of neutralization of TLR2 along with either TNF-α or IL-1β on amelioration of arthritis correlating with mTOR/STAT3 expression. Contribution of T lymphocytes collected from blood, spleen, synovial tissues, their derived cytokines IFN-γ, IL-6, IL-17, TGF-β, IL-10 were noted. Expression of TLR2, TNFR1, TNFR2, NF-κB along with mTOR/STAT3 also recorded. Neutralization of TLR2 along with TNF-α and IL-1β were able to shift Th17 cells into immunosuppressive Treg cells. Furthermore,elevated expression of IL-10, TNFR2 and demoted expression of mTOR/ STAT3 along with NF-κB in lymphocytes confirms its role in resolution of arthritis. It was also effective in reducing oxidative stress via increasing expression of the antioxidant enzymes. As a result, it can be inferred that Treg-derived IL-10, which may mitigate inflammatory effects of septic arthritis by influencing the mTOR/STAT3 interaction in lymphocytes, may be selected as a different therapeutic strategy for reducing the impact of septic arthritis.
Collapse
Affiliation(s)
- Rituparna Ghosh
- Department of Physiology, Immunology laboratory, University of Calcutta, University Colleges of Science and Technology, 92 APC Road, Calcutta 700009, West Bengal, India.
| | - Biswadev Bishayi
- Department of Physiology, Immunology laboratory, University of Calcutta, University Colleges of Science and Technology, 92 APC Road, Calcutta 700009, West Bengal, India.
| |
Collapse
|
3
|
Chauhan S, Nusbaum RJ, Huante MB, Holloway AJ, Endsley MA, Gelman BB, Lisinicchia JG, Endsley JJ. Therapeutic Modulation of Arginase with nor-NOHA Alters Immune Responses in Experimental Mouse Models of Pulmonary Tuberculosis including in the Setting of Human Immunodeficiency Virus (HIV) Co-Infection. Trop Med Infect Dis 2024; 9:129. [PMID: 38922041 PMCID: PMC11209148 DOI: 10.3390/tropicalmed9060129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/28/2024] [Accepted: 06/03/2024] [Indexed: 06/27/2024] Open
Abstract
L-arginine metabolism is strongly linked with immunity to mycobacteria, primarily through the antimicrobial activity of nitric oxide (NO). The potential to modulate tuberculosis (TB) outcomes through interventions that target L-arginine pathways are limited by an incomplete understanding of mechanisms and inadequate in vivo modeling. These gaps in knowledge are compounded for HIV and Mtb co-infections, where activation of arginase-1 due to HIV infection may promote survival and replication of both Mtb and HIV. We utilized in vitro and in vivo systems to determine how arginase inhibition using Nω-hydroxy-nor-L-arginine (nor-NOHA) alters L-arginine pathway metabolism relative to immune responses and disease outcomes following Mtb infection. Treatment with nor-NOHA polarized murine macrophages (RAW 264.7) towards M1 phenotype, increased NO, and reduced Mtb in RAW macrophages. In Balb/c mice, nor-NOHA reduced pulmonary arginase and increased the antimicrobial metabolite spermine in association with a trend towards reduced Mtb CFU in lung. In humanized immune system (HIS) mice, HIV infection increased plasma arginase and heightened the pulmonary arginase response to Mtb. Treatment with nor-NOHA increased cytokine responses to Mtb and Mtb/HIV in lung tissue but did not significantly alter bacterial burden or viral load. Our results suggest that L-arginine pathway modulators may have potential as host-directed therapies to augment antibiotics in TB chemotherapy.
Collapse
Affiliation(s)
- Sadhana Chauhan
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (S.C.); (R.J.N.); (M.B.H.); (A.J.H.); (M.A.E.)
| | - Rebecca J. Nusbaum
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (S.C.); (R.J.N.); (M.B.H.); (A.J.H.); (M.A.E.)
| | - Matthew B. Huante
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (S.C.); (R.J.N.); (M.B.H.); (A.J.H.); (M.A.E.)
| | - Alex J. Holloway
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (S.C.); (R.J.N.); (M.B.H.); (A.J.H.); (M.A.E.)
| | - Mark A. Endsley
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (S.C.); (R.J.N.); (M.B.H.); (A.J.H.); (M.A.E.)
| | - Benjamin B. Gelman
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; (B.B.G.); (J.G.L.)
| | - Joshua G. Lisinicchia
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; (B.B.G.); (J.G.L.)
| | - Janice J. Endsley
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (S.C.); (R.J.N.); (M.B.H.); (A.J.H.); (M.A.E.)
| |
Collapse
|
4
|
Roy S, Roy S, Halder S, Jana K, Ukil A. Leishmania exploits host cAMP/EPAC/calcineurin signaling to induce an IL-33-mediated anti-inflammatory environment for the establishment of infection. J Biol Chem 2024; 300:107366. [PMID: 38750790 PMCID: PMC11208913 DOI: 10.1016/j.jbc.2024.107366] [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: 12/01/2023] [Revised: 05/03/2024] [Accepted: 05/05/2024] [Indexed: 06/10/2024] Open
Abstract
Host anti-inflammatory responses are critical for the progression of visceral leishmaniasis, and the pleiotropic cytokine interleukin (IL)-33 was found to be upregulated in infection. Here, we documented that IL-33 induction is a consequence of elevated cAMP-mediated exchange protein activated by cAMP (EPAC)/calcineurin-dependent signaling and essential for the sustenance of infection. Leishmania donovani-infected macrophages showed upregulation of IL-33 and its neutralization resulted in decreased parasite survival and increased inflammatory responses. Infection-induced cAMP was involved in IL-33 production and of its downstream effectors PKA and EPAC, only the latter was responsible for elevated IL-33 level. EPAC initiated Rap-dependent phospholipase C activation, which triggered the release of intracellular calcium followed by calcium/calmodulin complex formation. Screening of calmodulin-dependent enzymes affirmed involvement of the phosphatase calcineurin in cAMP/EPAC/calcium/calmodulin signaling-induced IL-33 production and parasite survival. Activated calcineurin ensured nuclear localization of the transcription factors, nuclear factor of activated T cell 1 and hypoxia-inducible factor 1 alpha required for IL-33 transcription, and we further confirmed this by chromatin immunoprecipitation assay. Administering specific inhibitors of nuclear factor of activated T cell 1 and hypoxia-inducible factor 1 alpha in BALB/c mouse model of visceral leishmaniasis decreased liver and spleen parasite burden along with reduction in IL-33 level. Splenocyte supernatants of inhibitor-treated infected mice further documented an increase in tumor necrosis factor alpha and IL-12 level with simultaneous decrease of IL-10, thereby indicating an overall disease-escalating effect of IL-33. Thus, this study demonstrates that cAMP/EPAC/calcineurin signaling is crucial for the activation of IL-33 and in effect creates anti-inflammatory responses, essential for infection.
Collapse
Affiliation(s)
- Souravi Roy
- Department of Biochemistry, University of Calcutta, Kolkata, India
| | - Shalini Roy
- Department of Biochemistry, University of Calcutta, Kolkata, India
| | - Satyajit Halder
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Kuladip Jana
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Anindita Ukil
- Department of Biochemistry, University of Calcutta, Kolkata, India.
| |
Collapse
|
5
|
de Franca MNF, Rodrigues LS, Barreto AS, da Cruz GS, Aragão-Santos JC, da Silva AM, de Jesus AR, Palatnik-de-Sousa CB, de Almeida RP, Corrêa CB. CD4 + Th1 and Th17 responses and multifunctional CD8 T lymphocytes associated with cure or disease worsening in human visceral leishmaniasis. Front Immunol 2024; 15:1277557. [PMID: 38410517 PMCID: PMC10895669 DOI: 10.3389/fimmu.2024.1277557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 01/02/2024] [Indexed: 02/28/2024] Open
Abstract
Introduction In VL, a proinflammatory phenotype is typically associated with enhanced phagocytosis and a Th1 mediated immune response resulting in infection control. In contrast, an anti-inflammatory phenotype, associated with a predominant regulatory response, typically enables intracellular multiplication of Leishmania parasites and disease progression. Methods To investigate the impact of chemotherapy on Th2 and Th17 immune responses in patients with visceral leishmaniasis (VL), we assessed all combinations of intracellular expression of IFN-γ, IL-10, IL-4 and IL-17 in the CD4+ and CD8+ T cell populations of peripheral blood mononuclear cell (PBMC) samples from patients, after antigenic stimulation with Leishmania lysate, throughout treatment and follow-up. As increases in spleen and liver sizes and decreases in hematocrit, hemogloblin, erythrocytes, monocytes, leukocytes and platelets levels are strongly related to the disease, we studied the correlations between the frequencies of T cells producing the afore mentioned cytokines, individually and in combination, and these variables, as markers of disease or cure. Results We found that the frequency of IFN-γ-producingCD4+ T cells increased until the end of chemotherapy with Glucantime® or AmBisome ®, while IL-10, IL-4 and IL-17-producing CD4+ T cells peaked on day 7 following the start of treatment. Although the frequency of CD4+IL-17+ cells decreased during treatment an increase was observed after clinical cure. The frequency of CD4+ T cells producing only IFN-γ or IL-17 correlated with blood monocytes levels. Frequencies of double-producers of IFN-γ and IL-10 or IL-4 correlated positively with eosinophils and platelets levels. Together, this suggest that IFN-γ drives the immune response towards Th1 at cure. In contrast, and associated with disease or Th2 response, the frequency of CD4+ IL-10+ cells correlated positively with spleen sizes and negatively with circulating monocyte levels, while the frequency of CD4+ producing both IL-4 and IL-10 correlated negatively with platelets levels. The frequency of CD8+ single-producers of IFN-γ increased from day 21 to 90 while that of single-producers of IL-10 peaked on day 7, of IL-4 on day 30 and of IL-17, on day 180. IFN-γ expression in CD8+ single- and double-producers of cytokines was indicative of an immune response associated with cure. In contrast, frequencies of CD8+ double-producers of IL-4 and IL-10, IL-4 and IL-17 and IL-10 and IL-17 and producers of three and four cytokines, were associated with disease and were low after the cure. Frequencies of CD8+ T cells producing IFN-γ alone or with IL-17 were positively correlated with platelets levels. In contrast, as markers of disease: 1) frequencies of single producers of IL-10 correlated negatively with leukocytes levels, 2) frequencies of double producers of IL-4 and IL-10 correlated negatively with platelet, leukocyte, lymphocyte and circulating monocyte levels, 3) frequencies of triple-producers of IFN-γ, IL-4 and IL-10 correlated negatively with platelet, leukocyte and neutrophil levels and 4) frequencies of producers of IFN-γ, IL-4, IL-10 and IL-17 simultaneously correlated positively with spleen size, and negatively with leukocyte and neutrophil levels. Discussion Our results confirmed that the clinical improvement of VL patients correlates with the decrease of an IL-4 and IL-10 CD4+Th2 response, the recovery of CD4+ Th1 and Th17 responses and the frequency of CD8+ single-producers of IFN-γ and double producers of IFN-γ and IL-17.
Collapse
Affiliation(s)
- Mariana Nobre Farias de Franca
- Laboratory of Biology and Immunology of Cancer and Leishmania, Department of Morphology, Federal University of Sergipe, Aracaju, Sergipe, Brazil
- Graduate Program in Health Sciences, Federal University of Sergipe, Aracaju, Sergipe, Brazil
| | - Lorranny Santana Rodrigues
- Laboratory of Biology and Immunology of Cancer and Leishmania, Department of Morphology, Federal University of Sergipe, Aracaju, Sergipe, Brazil
- Graduate Program in Health Sciences, Federal University of Sergipe, Aracaju, Sergipe, Brazil
| | - Aline Silva Barreto
- Graduate Program in Health Sciences, Federal University of Sergipe, Aracaju, Sergipe, Brazil
- Division of Immunology and Molecular Biology Laboratory, University Hospital/Empresa Brasileira de Serviços Hospitalares (EBSERBH), Federal University of Sergipe, Aracaju, Sergipe, Brazil
| | - Geydson Silveira da Cruz
- Graduate Program in Health Sciences, Federal University of Sergipe, Aracaju, Sergipe, Brazil
- Division of Immunology and Molecular Biology Laboratory, University Hospital/Empresa Brasileira de Serviços Hospitalares (EBSERBH), Federal University of Sergipe, Aracaju, Sergipe, Brazil
| | - José Carlos Aragão-Santos
- Graduate Program in Health Sciences, Federal University of Sergipe, Aracaju, Sergipe, Brazil
- Department of Physical Education, Federal University of Sergipe, Aracaju, Sergipe, Brazil
| | - Angela Maria da Silva
- Graduate Program in Health Sciences, Federal University of Sergipe, Aracaju, Sergipe, Brazil
- Department of Medicine, Federal University of Sergipe, Aracaju, Sergipe, Brazil
| | - Amélia Ribeiro de Jesus
- Graduate Program in Health Sciences, Federal University of Sergipe, Aracaju, Sergipe, Brazil
- Division of Immunology and Molecular Biology Laboratory, University Hospital/Empresa Brasileira de Serviços Hospitalares (EBSERBH), Federal University of Sergipe, Aracaju, Sergipe, Brazil
- Department of Medicine, Federal University of Sergipe, Immunology Investigative Institute (III), INCT, CNPq, Aracaju, Sergipe, Brazil
| | - Clarisa B. Palatnik-de-Sousa
- Graduate Program in Health Sciences, Federal University of Sergipe, Aracaju, Sergipe, Brazil
- Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Immunology Investigative Institute (III), Insititutos nacionais de Ciência e Tecnologia (INCT), Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq), Rio de Janeiro, Brazil
- Graduate Program in Vegetal Biotechnology and Bioprocesses, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Roque Pacheco de Almeida
- Graduate Program in Health Sciences, Federal University of Sergipe, Aracaju, Sergipe, Brazil
- Division of Immunology and Molecular Biology Laboratory, University Hospital/Empresa Brasileira de Serviços Hospitalares (EBSERBH), Federal University of Sergipe, Aracaju, Sergipe, Brazil
- Department of Medicine, Federal University of Sergipe, Immunology Investigative Institute (III), INCT, CNPq, Aracaju, Sergipe, Brazil
| | - Cristiane Bani Corrêa
- Laboratory of Biology and Immunology of Cancer and Leishmania, Department of Morphology, Federal University of Sergipe, Aracaju, Sergipe, Brazil
- Graduate Program in Health Sciences, Federal University of Sergipe, Aracaju, Sergipe, Brazil
| |
Collapse
|
6
|
Devender M, Sebastian P, Maurya VK, Kumar K, Anand A, Namdeo M, Maurya R. Immunogenicity and protective efficacy of tuzin protein as a vaccine candidate in Leishmania donovani-infected BALB/c mice. Front Immunol 2024; 14:1294397. [PMID: 38274802 PMCID: PMC10808571 DOI: 10.3389/fimmu.2023.1294397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 12/19/2023] [Indexed: 01/27/2024] Open
Abstract
Visceral leishmaniasis (VL) is referred to as the most severe and fatal type of leishmaniasis basically caused by Leishmania donovani and L. infantum. The most effective method for preventing the spread of the disease is vaccination. Till today, there is no promising licensed vaccination for human VL. Hence, investigation for vaccines is necessary to enrich the therapeutic repertoire against leishmaniasis. Tuzin is a rare trans-membrane protein that has been reported in Trypanosoma cruzi with unknown function. However, tuzin is not characterized in Leishmania parasites. In this study, we for the first time demonstrated that tuzin protein was expressed in both stages (promastigote and amastigote) of L. donovani parasites. In-silico studies revealed that tuzin has potent antigenic properties. Therefore, we analyzed the immunogenicity of tuzin protein and immune response in BALB/c mice challenged with the L. donovani parasite. We observed that tuzin-vaccinated mice have significantly reduced parasite burden in the spleen and liver compared with the control. The number of granulomas in the liver was also significantly decreased compared with the control groups. We further measured the IgG2a antibody level, a marker of Th1 immune response in VL, which was significantly higher in the serum of immunized mice when compared with the control. Splenocytes stimulated with soluble Leishmania antigen (SLA) displayed a significant increase in NO and ROS levels compared with the control groups. Tuzin-immunized and parasite-challenged mice exhibit a notable rise in the IFN-γ/IL-10 ratio by significantly suppressing IL-10 expression level, an immunosuppressive cytokine that inhibits leishmanicidal immune function and encourages disease progression. In conclusion, tuzin immunizations substantially increase the protective immune response in L. donovani-challenged mice groups compared with control.
Collapse
|
7
|
Nzoumbou-Boko R, Zolipou COOK, Yambiyo BM, Semballa S, Nalingbo MCIDM, Daulouède S, Vincendeau P. Optimization of the arginase activity assay micromethod for macrophages and sera. BMC Res Notes 2023; 16:188. [PMID: 37644583 PMCID: PMC10466829 DOI: 10.1186/s13104-023-06462-4] [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: 07/30/2022] [Accepted: 08/15/2023] [Indexed: 08/31/2023] Open
Abstract
OBJECTIVE We optimized the spectrophotometric micromethod for the determination of arginase activity based on the Corraliza et al. modification of Schimke's method. Arginase activity in sera from patients suffering from human African trypanosomiasis, in macrophage lysates from trypanosome-infected mice, and in purified bovine liver arginase was compared using the conventional and optimized micromethods. RESULTS The sensitivity of both micromethods was comparable. However, our optimized method has the following advantages: it uses small sample volumes (6 µl per assay vs. 50 µl) and reagent volumes (200 µl vs. 400 µl), it can be carried out in a single microplate well, thereby minimizing handling, and it requires fewer materials and utilizes readily available equipment. Our optimized method proved to be applicable and well suited for small-volume samples and resource-poor laboratories.
Collapse
Affiliation(s)
- Romaric Nzoumbou-Boko
- Laboratoire de Parasitologie, Institut Pasteur de Bangui, BP 923, Bangui, Central African Republic.
| | - Cyrille Oliver Ozzin-Kholy Zolipou
- Laboratoire de Parasitologie, Institut Pasteur de Bangui, BP 923, Bangui, Central African Republic
- Laboratoire de Parasitologie, UMR 177 IRD/CIRAD "INTERTRYP," Université Bordeaux, Bordeaux, F-33000, France
| | - Brice Martial Yambiyo
- Service d'Epidémiologie, Institut Pasteur de Bangui, BP 923, Bangui, Central African Republic
| | - Silla Semballa
- Laboratoire des Sciences Biologiques et Agronomiques pour le Développement (LASBAD), Université de Bangui, République Centrafricaine, Bangui, Central African Republic
| | | | - Sylvie Daulouède
- Laboratoire de Parasitologie, UMR 177 IRD/CIRAD "INTERTRYP," Université Bordeaux, Bordeaux, F-33000, France
| | - Philippe Vincendeau
- Laboratoire de Parasitologie, UMR 177 IRD/CIRAD "INTERTRYP," Université Bordeaux, Bordeaux, F-33000, France
| |
Collapse
|
8
|
Mohapatra S, Cafiero J, Kashfi K, Mehta P, Banerjee P. Why Don't the Mutant Cells That Evade DNA Repair Cause Cancer More Frequently? Importance of the Innate Immune System in the Tumor Microenvironment. Int J Mol Sci 2023; 24:5026. [PMID: 36902456 PMCID: PMC10002487 DOI: 10.3390/ijms24055026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/03/2023] [Accepted: 03/04/2023] [Indexed: 03/08/2023] Open
Abstract
The standard of care for most malignant solid tumors still involves tumor resection followed by chemo- and radiation therapy, hoping to eliminate the residual tumor cells. This strategy has been successful in extending the life of many cancer patients. Still, for primary glioblastoma (GBM), it has not controlled recurrence or increased the life expectancies of patients. Amid such disappointment, attempts to design therapies using the cells in the tumor microenvironment (TME) have gained ground. Such "immunotherapies" have so far overwhelmingly used genetic modifications of Tc cells (Car-T cell therapy) or blocking of proteins (PD-1 or PD-L1) that inhibit Tc-cell-mediated cancer cell elimination. Despite such advances, GBM has remained a "Kiss of Death" for most patients. Although the use of innate immune cells, such as the microglia, macrophages, and natural killer (NK) cells, has been considered in designing therapies for cancers, such attempts have not reached the clinic yet. We have reported a series of preclinical studies highlighting strategies to "re-educate" GBM-associated microglia and macrophages (TAMs) so that they assume a tumoricidal status. Such cells then secrete chemokines to recruit activated, GBM-eliminating NK cells and cause the rescue of 50-60% GBM mice in a syngeneic model of GBM. This review discusses a more fundamental question that most biochemists harbor: "since we are generating mutant cells in our body all the time, why don't we get cancer more often?" The review visits publications addressing this question and discusses some published strategies for re-educating the TAMs to take on the "sentry" role they initially maintained in the absence of cancer.
Collapse
Affiliation(s)
- Shubhasmita Mohapatra
- Department of Chemistry, The College of Staten Island, City University of New York, Staten Island, NY 10314, USA
| | - Jared Cafiero
- Department of Chemistry, The College of Staten Island, City University of New York, Staten Island, NY 10314, USA
| | - Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY 10031, USA
- Graduate Program in Biology, City University of New York Graduate Center, New York, NY 10016, USA
| | - Parag Mehta
- Aveta Biomics, Inc., 110 Great Road, Suite 302, Bedford, MA 01730, USA
| | - Probal Banerjee
- Department of Chemistry, The College of Staten Island, City University of New York, Staten Island, NY 10314, USA
- Graduate Program in Biology, City University of New York Graduate Center, New York, NY 10016, USA
| |
Collapse
|
9
|
Leishmanicidal Activity of Guanidine Derivatives against Leishmania infantum. Trop Med Infect Dis 2023; 8:tropicalmed8030141. [PMID: 36977142 PMCID: PMC10051705 DOI: 10.3390/tropicalmed8030141] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/19/2023] [Accepted: 02/17/2023] [Indexed: 03/02/2023] Open
Abstract
Leishmaniasis is a neglected tropical infectious disease with thousands of cases annually; it is of great concern to global health, particularly the most severe form, visceral leishmaniasis. Visceral leishmaniasis treatments are minimal and have severe adverse effects. As guanidine-bearing compounds have shown antimicrobial activity, we analyzed the cytotoxic effects of several guanidine-bearing compounds on Leishmania infantum in their promastigote and amastigote forms in vitro, their cytotoxicity in human cells, and their impact on reactive nitrogen species production. LQOFG-2, LQOFG-6, and LQOFG-7 had IC50 values of 12.7, 24.4, and 23.6 µM, respectively, in promastigotes. These compounds exhibited cytotoxicity in axenic amastigotes at 26.1, 21.1, and 18.6 µM, respectively. The compounds showed no apparent cytotoxicity in cells from healthy donors. To identify mechanisms of action, we evaluated cell death processes by annexin V and propidium iodide staining and nitrite production. Guanidine-containing compounds caused a significant percentage of death by apoptosis in amastigotes. Independent of L. infantum infection, LQOFG-7 increased nitrite production in peripheral blood mononuclear cells, which suggests a potential mechanism of action for this compound. Therefore, these data suggest that guanidine derivatives are potential anti-microbial molecules, and further research is needed to fully understand their mechanism of action, especially in anti-leishmanial studies.
Collapse
|
10
|
Almeida-Souza F, da Silva VD, Taniwaki NN, Hardoim DDJ, Mendonça Filho AR, Moreira WFDF, Buarque CD, Calabrese KDS, Abreu-Silva AL. Nitric Oxide Induction in Peritoneal Macrophages by a 1,2,3-Triazole Derivative Improves Its Efficacy upon Leishmania amazonensis In Vitro Infection. J Med Chem 2021; 64:12691-12704. [PMID: 34427442 DOI: 10.1021/acs.jmedchem.1c00725] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
1,2,3-Triazole is one of the most flexible chemical scaffolds broadly used in various fields. Here, we report the antileishmanial activity of 1,2,3-triazole derivatives, the ultrastructural alterations induced by their treatment, and the nitric oxide (NO) modulation effect on their efficacy against Leishmania amazonensis in vitro infection. After the screening of eleven compounds, compound 4 exhibited better results against L. amazonensis promastigotes (IC50 = 15.52 ± 3.782 μM) and intracellular amastigotes (IC50 = 4.10 ± 1.136 μM), 50% cytotoxicity concentration at 84.01 ± 3.064 μM against BALB/c peritoneal macrophages, and 20.49-fold selectivity for the parasite over the cells. Compound 4 induced ultrastructural mitochondrial alterations and lipid inclusions in L. amazonensis promastigotes, upregulated tumor necrosis factor α, interleukin (IL)-1β, IL-6, IL-12, and IL-10 messenger RNA expressions, and enhanced the NO production, verified by nitrite (p = 0.0095) and inducible nitric oxide synthase expression (p = 0.0049) quantification, which played an important role in its activity against intramacrophagic L. amazonensis. In silico prediction in association with antileishmanial activity results showed compound 4 as a hit compound with promising potential for further studies of new leishmaniasis treatment options.
Collapse
Affiliation(s)
- Fernando Almeida-Souza
- Laboratório de Anatomopatologia, Departamento de Patologia, Universidade Estadual do Maranhão, 65055-310 São Luís, Maranhão, Brazil.,Laboratório de Imunomodulação e Protozoologia, Instituto Oswaldo Cruz, Fiocruz, 21040-900 Rio de Janeiro, Brazil
| | - Verônica Diniz da Silva
- Laboratório de Síntese Orgânica, Pontifícia Universidade Católica, 22451-900 Rio de Janeiro, Rio de Janeiro, Brazil
| | - Noemi Nosomi Taniwaki
- Núcleo de Microscopia Eletrônica, Instituto Adolfo Lutz, 01246-000 São Paulo, São Paulo, Brazil
| | - Daiana de Jesus Hardoim
- Laboratório de Imunomodulação e Protozoologia, Instituto Oswaldo Cruz, Fiocruz, 21040-900 Rio de Janeiro, Brazil
| | - Ailésio Rocha Mendonça Filho
- Laboratório de Anatomopatologia, Departamento de Patologia, Universidade Estadual do Maranhão, 65055-310 São Luís, Maranhão, Brazil
| | | | - Camilla Djenne Buarque
- Laboratório de Síntese Orgânica, Pontifícia Universidade Católica, 22451-900 Rio de Janeiro, Rio de Janeiro, Brazil
| | - Kátia da Silva Calabrese
- Laboratório de Imunomodulação e Protozoologia, Instituto Oswaldo Cruz, Fiocruz, 21040-900 Rio de Janeiro, Brazil
| | - Ana Lucia Abreu-Silva
- Laboratório de Anatomopatologia, Departamento de Patologia, Universidade Estadual do Maranhão, 65055-310 São Luís, Maranhão, Brazil
| |
Collapse
|