1
|
Ly AT, Diop D, Diop M, Schacht AM, Mbengue A, Diagne R, Guisse M, Dompnier JP, Messias C, Coler RN, Ramos CR, Tendeng JN, Ndiaye S, Marroquin-Quelopana M, de Carvalho Parra J, Dos Santos T, Sirianni Dos Santos Almeida M, Mendes-da-Cruz DA, Reed S, Savino W, Riveau G, Tendler M. The Sm14+GLA-SE Recombinant Vaccine Against Schistosoma mansoni and S. haematobium in Adults and School Children: Phase II Clinical Trials in West Africa. Vaccines (Basel) 2025; 13:316. [PMID: 40266229 DOI: 10.3390/vaccines13030316] [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: 01/15/2025] [Revised: 02/10/2025] [Accepted: 02/27/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND/OBJECTIVES Following previous successful Phase I clinical trials conducted in men and women in a non-endemic area for schistosomiasis in Brazil, the Sm14 vaccine was evaluated in an endemic region in Senegal. We report successful clinical trials in adults (Phase IIa) and school children (Phase IIb), respectively, of a Schistosoma mansoni 14 kDa fatty acid-binding protein (Sm14) vaccine + a glucopyranosyl lipid A (GLA-SE) adjuvant. METHODS Participants were evaluated based on clinical assessments, laboratory tests (including hematologic and biochemical analyses of renal and hepatic functions), and immunological parameters (humoral and cellular responses) up to 12 months after the first vaccination dose in the Phase IIa trial and after 120 days in the Phase IIb trial. RESULTS The results showed strong immunogenic responses and good tolerance in both adults and children, with no major adverse effects. Importantly, significant increases in Sm14-specific total IgG (IgG1 and IgG3) were observed as early as 30 days after the first vaccination, with high titres remaining at least 120 days afterwards. Sm14-specific total IgG serum levels were also significantly enhanced in adults and in both infected and non-infected, vaccinated children and elicited robust cytokine responses with increased TNFα, IFN-γ, and IL-2 profiles. CONCLUSIONS Overall, the Sm14+GLA-SE vaccine is safe and highly immunogenic, with a clearly protective potential against schistosomiasis, supporting progression to the next Phase III clinical trials.
Collapse
Affiliation(s)
- Amadou Tidjani Ly
- Biomedical Research Center Espoir Pour La Santé, Saint Louis BP 226, Senegal
| | - Doudou Diop
- Biomedical Research Center Espoir Pour La Santé, Saint Louis BP 226, Senegal
| | - Modou Diop
- Biomedical Research Center Espoir Pour La Santé, Saint Louis BP 226, Senegal
| | - Anne-Marie Schacht
- Biomedical Research Center Espoir Pour La Santé, Saint Louis BP 226, Senegal
- CIIL-Center for Infection and Immunity of Lille, Institut Pasteur de Lille, University of Lille, CNRS UMR, Inserm U1019-Lille, F-59000 Lille, France
| | - Abdoulaye Mbengue
- Biomedical Research Center Espoir Pour La Santé, Saint Louis BP 226, Senegal
| | - Rokhaya Diagne
- Biomedical Research Center Espoir Pour La Santé, Saint Louis BP 226, Senegal
| | - Marieme Guisse
- Biomedical Research Center Espoir Pour La Santé, Saint Louis BP 226, Senegal
| | | | - Carolina Messias
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil
- Brazilian National Institute of Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil
| | - Rhea N Coler
- Seattle Children's Research Institute, Center for Global Infectious Disease Research, Seattle, WA 98105, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98105, USA
- Department of Global Health, University of Washington, Seattle, WA 98105, USA
| | - Celso R Ramos
- Fenix Biotec Treinamento SS LTDA, São Paulo 05591-090, Brazil
| | - Jacques-Noël Tendeng
- Hopital Régional de Saint Louis, UFR 2S, Université Gaston Berger, Saint Louis BP 226, Senegal
| | - Seynabou Ndiaye
- Région Médicale de Saint Louis, Ministère de la Santé et de l'Action Sociale, Saint Louis BP 226, Senegal
| | | | - Juçara de Carvalho Parra
- Laboratory of Anti-Helminth Vaccine Research and Development, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21045-900, Brazil
| | - Tatiane Dos Santos
- Laboratory of Anti-Helminth Vaccine Research and Development, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21045-900, Brazil
| | - Marília Sirianni Dos Santos Almeida
- Laboratory of Anti-Helminth Vaccine Research and Development, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21045-900, Brazil
| | - Daniella Arêas Mendes-da-Cruz
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil
- Brazilian National Institute of Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil
- Rio de Janeiro Research Network on Neuroinflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil
- INOVA-IOC Network on Neuroimmunomodulation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil
| | | | - Wilson Savino
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil
- Brazilian National Institute of Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil
- Rio de Janeiro Research Network on Neuroinflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil
- INOVA-IOC Network on Neuroimmunomodulation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil
| | - Gilles Riveau
- Biomedical Research Center Espoir Pour La Santé, Saint Louis BP 226, Senegal
- CIIL-Center for Infection and Immunity of Lille, Institut Pasteur de Lille, University of Lille, CNRS UMR, Inserm U1019-Lille, F-59000 Lille, France
| | - Miriam Tendler
- Laboratory of Anti-Helminth Vaccine Research and Development, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21045-900, Brazil
- FABP Biotech, Rio de Janeiro 22611-100, Brazil
| |
Collapse
|
2
|
Silva-Freitas ML, Corrêa-Castro G, Da-Cruz AM, Santos-Oliveira JR. Insights to the HIV-associated visceral leishmaniasis clinical outcome: lessons learned about immune mediated disorders. Front Immunol 2025; 16:1516176. [PMID: 40145085 PMCID: PMC11937021 DOI: 10.3389/fimmu.2025.1516176] [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: 10/23/2024] [Accepted: 02/19/2025] [Indexed: 03/28/2025] Open
Abstract
Most cases of visceral leishmaniasis (VL) and human immunodeficiency virus (HIV) co-infection (VL/HIV) in the Americas occur in Brazil, and the prevalence of VL/HIV has been increasing since 2019, reaching 19% in 2023. This association presents a challenge for the management of VL, since both VL and HIV infection share immunopathogenic characteristics that can reciprocally affect co-infected patients. Thus, VL may contribute to the immunosuppression and other immunological disturbances associated with the rapid progression to acquired immunodeficiency syndrome (AIDS), whereas HIV infection accelerates the development of active VL and reduces the probability of a successful response to anti-Leishmania therapy, resulting in an increase in the relapse and lethality rates of VL. In this synergistic impairment, one of the most critical hallmarks of VL/HIV co-infection is the enhancement of immunosuppression and intense chronic immune activation, caused not only by each infection per se, but also by the cytokine storm and translocation of microbial products. Thus, co-infected patients present with an impaired effector immune response that may result in inefficient parasitic control. In addition, the chronic activation environment in VL/HIV patients may favor progression to early immunosenescence and exhaustion, worsening the patients' clinical condition and increasing the frequency of disease relapse. Herein, we review the immunological parameters associated with the immunopathogenesis of VL/HIV co-infection that could serve as good biomarkers of clinical prognosis in terms of relapse and severity of VL.
Collapse
Affiliation(s)
- Maria Luciana Silva-Freitas
- Laboratório Interdisciplinar de Pesquisas Médicas - Instituto Oswaldo Cruz – Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Gabriela Corrêa-Castro
- Laboratório Interdisciplinar de Pesquisas Médicas - Instituto Oswaldo Cruz – Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Alda Maria Da-Cruz
- Laboratório Interdisciplinar de Pesquisas Médicas - Instituto Oswaldo Cruz – Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
- Departamento de Microbiologia, Immunologia e Parasitologia (DMIP), Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro, Brazil
- Instituto Nacional de Ciência, Tecnologia e Inovação - Neuroimunomodulação (INCT - NIM), Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq), Brasília, Brazil
- Departamento de Doenças Transmissíveis, Secretaria de Vigilância em Saúde e Ambiente, Ministério da Saúde, Brasília, Brazil
| | - Joanna Reis Santos-Oliveira
- Laboratório Interdisciplinar de Pesquisas Médicas - Instituto Oswaldo Cruz – Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
- Instituto Nacional de Ciência, Tecnologia e Inovação - Neuroimunomodulação (INCT - NIM), Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq), Brasília, Brazil
- Núcleo de Ciências Biomédicas Aplicadas, Instituto Federal de Educação, Ciência e Tecnologia do Rio de Janeiro (IFRJ), Rio de Janeiro, Brazil
| |
Collapse
|
3
|
de Oliveira Mendes-Aguiar C, do Monte Alves M, Lopes Machado ADA, Monteiro GRDG, Marques Medeiros I, Queiroz JW, Lima ID, Costa EG, Pearson RD, Wilson ME, Glesby MJ, do Nascimento ELT, Jerônimo SMB. T-cell activation and senescence in asymptomatic HIV/Leishmania infantum co-infection. PLoS Negl Trop Dis 2025; 19:e0012848. [PMID: 40096173 PMCID: PMC11964262 DOI: 10.1371/journal.pntd.0012848] [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: 02/19/2024] [Revised: 04/02/2025] [Accepted: 01/16/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Leishmania infantum can be an opportunistic pathogen, with an immunocompromised status increasing the risk of converting asymptomatic infection to symptomatic visceral leishmaniasis (VL). VL has approximately 5% fatality rate; and HIV coinfection (AIDS/VL) increases this risk. We hypothesized that, relative to those with HIV alone, people with co-infection would have altered T cell activation which could impact on the risk of VL. METHODS A cross-sectional study was performed between 2014 and 2016 to determine the prevalence of L. infantum infection in people living with HIV (PLHIV) residing in Brazil (n = 1,372). Subsequent incident cases of VL were ascertained from a public health database through 2018 and from a cohort of families with VL. Immune status of 69 participants was evaluated and comparisons made between those with and without HIV, with latent or with active Leishmania infection and those without HIV but with active or resolved Leishmania or T cell hypersensitivity to Leishmania antigen and healthy control subjects. RESULTS A total of 24.2% of PLHIV had positive anti-IgG L. infantum antibodies. The relative risk of developing AIDS/VL was 2.27 (95% CI: 0.920 to 5.59; p = 0.07) to HIV/Leish coinfected subjects with positive leishmania serology compared to HIV subjects without leishmania serology. Poor adherence to antiretroviral therapy (p = 0.0008) or prior opportunistic infections (p = 0.0007) was associated with development of AIDS/VL in asymptomatic HIV/Leish. CD4+ and CD8+ T cells counts or viral load were similar between asymptomatic HIV/Leish and HIV subjects. However, activated CD8+CD38+HLA-DR+ T cells were higher in asymptomatic HIV/Leish than HIV. Likewise, senescent (CD57+) and PD1+ CD8+ T cells were higher in asymptomatic HIV/Leish than in AIDS/VL or HIV groups. CONCLUSION Although asymptomatic HIV/Leish subjects had CD4+ and CD8+ T cells similar to HIV alone, their CD8+T cells had increased activation and senescence which could contribute to risk of developing VL.
Collapse
Affiliation(s)
| | - Manoella do Monte Alves
- Department of Infectious Disease, Health Science Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | | | | | - Iara Marques Medeiros
- Institute of Tropical Medicine of Rio Grande do Norte, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Jose Wilton Queiroz
- Institute of Tropical Medicine of Rio Grande do Norte, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Iraci Duarte Lima
- Institute of Tropical Medicine of Rio Grande do Norte, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Eliardo G. Costa
- Department of Statistics, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Richard D. Pearson
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, United States of America
| | - Mary E. Wilson
- Departments of Internal Medicine and Microbiology & Immunology, University of Iowa and the Veterans’ Affairs Medical Center, Iowa City, Iowa, United States of America
| | - Marshall J. Glesby
- Division of Infectious Disease, Weill Cornell Medical College, New York, New York, United States of America
| | - Eliana Lúcia Tomaz do Nascimento
- Institute of Tropical Medicine of Rio Grande do Norte, Federal University of Rio Grande do Norte, Natal, Brazil
- Department of Infectious Disease, Health Science Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Selma Maria Bezerra Jerônimo
- Institute of Tropical Medicine of Rio Grande do Norte, Federal University of Rio Grande do Norte, Natal, Brazil
- Institute of Science and Technology of Tropical Diseases, Natal, Brazil
- Department of Biochemistry, Federal University of Rio Grande do Norte, Natal, Brazil
| |
Collapse
|
4
|
Itakura T, Sasaki H, Hosoya T, Umezawa N, Saito T, Iwai H, Hasegawa H, Sato H, Hirakawa A, Imai K, Morio T, Kimura N, Yasuda S. The role of TRECs/KRECs as immune indicators that reflect immunophenotypes and predict the risk of infection in systemic autoimmune diseases. Immunol Med 2025:1-12. [PMID: 39895338 DOI: 10.1080/25785826.2025.2460275] [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: 11/11/2024] [Accepted: 01/17/2025] [Indexed: 02/04/2025] Open
Abstract
T cell receptor rearrangement excision circles (TRECs) and immunoglobulin κ-deleting recombination excision circles (KRECs) represent the lymphopoiesis capacity, widely used for newborn screening of inborn errors of immunity. To clarify the significance of TRECs and KRECs as immune indicators in patients with systemic autoimmune diseases, we prospectively evaluated TREC and KREC levels with qPCR, lymphocyte phenotypes with flow cytometry, along with lymphocyte counts and serum immunoglobulin levels in peripheral blood samples from newly diagnosed patients. Each variable was assessed before immunosuppressive treatments (baseline), 3-, 6-, and 12-months after the treatment. Severe infections were recorded until 6 months after treatment. Among 35 patients, TREC and KREC levels were associated positively with the proportion of recent thymic emigrants, naïve T and B cells at all the timepoints. TREC and KREC levels decreased after treatment. The ratios of TREC and KREC levels under treatment to baseline were significantly lower in patients with severe infection than those without. In conclusion, TREC and KREC levels reflect peripheral blood immunophenotypes, specifically recent-emigrated T and B cells, in patients under treatment-naïve and immunosuppressive conditions. The longitudinal changes in TREC and KREC levels were beneficial markers for predicting the risk of severe infection during immunosuppressive treatments.
Collapse
Affiliation(s)
- Takuji Itakura
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Hirokazu Sasaki
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Tadashi Hosoya
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Natsuka Umezawa
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Tetsuya Saito
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Hideyuki Iwai
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | | | - Hiroyuki Sato
- Department of Clinical Biostatistics, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Akihiro Hirakawa
- Department of Clinical Biostatistics, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Kohsuke Imai
- Department of Pediatrics, National Defense Medical College, Tokorozawa, Japan
| | - Tomohiro Morio
- Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Naoki Kimura
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Shinsuke Yasuda
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| |
Collapse
|
5
|
Dhulipalla M, Chouhan G. The nexus between Leishmania & HIV: Debilitating host immunity and Hastening Comorbid disease burden. Exp Parasitol 2024; 265:108826. [PMID: 39147120 DOI: 10.1016/j.exppara.2024.108826] [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: 04/17/2024] [Revised: 07/28/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
The scintillating association between Leishmania and HIV has contributed exceptionally towards expansion of Visceral Leishmaniasis (VL) with Acquired Immunodeficiency Syndrome (AIDS). The co-infection poses a grievous threat to elimination of VL and containment of Human Immunodeficiency Virus (HIV). When coinfected, Leishmania and HIV complement each other's proliferation and survival by inducing immunesenescence, T cell fatigue and exhaustion. Antigen presentation is lost, co-stimulatory molecules are diminished whereas co-inhibitory molecules such as CTLA-4, TIGIT, LAG-3 etc. are upregulated to ensure a Th2-baised immune environment. As a consequence, Leishmania-HIV coinfection causes poor outcomes, inflates the spread of Leishmania parasites, enhances the severity of side-effects to drugs, as well as escalate the probability of treatment failure and mortality. What makes control extremely strenuous is that there are frequent episodes of VL relapse with no prognostic markers, no standard immunophenotype(s) and appearance of atypical clinical symptoms. Thus, a standard therapeutic regimen has been difficult to develop and treatment is majorly dependent upon a combination of liposomal Amphotericin B and Miltefosine, a therapy that is expensive and capable of causing drastic side-effects in recipients. As World Health Organization is committed to eliminate both VL and HIV in due course of future, the existing therapeutic interventions require advancements to grapple and overcome this hazardous co-infection. In this context, an overview of HIV-VL co-infection, immunopathology of HIV and Leishmania co-inhabitance, available therapeutic options and their limitations in the treatment of co-infection are discussed in-depth.
Collapse
Affiliation(s)
- Manasvi Dhulipalla
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, 201306, India
| | - Garima Chouhan
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, 201306, India.
| |
Collapse
|
6
|
Silva RVS, Uliana SRB, Yasunaka JKUY, Veloso CS, Sousa E, Ferreira MML, Carvalho VS, Ferreira GR, Costa DL, Costa CHN. Low Plasma Lipids Are Associated with Relapsing and Lethal Visceral Leishmaniasis in HIV-Infected Patients. Pathogens 2024; 13:450. [PMID: 38921748 PMCID: PMC11206293 DOI: 10.3390/pathogens13060450] [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: 04/28/2024] [Revised: 05/18/2024] [Accepted: 05/23/2024] [Indexed: 06/27/2024] Open
Abstract
Visceral leishmaniasis (VL) results from protozoa Leishmania infantum and L. donovani infection. This study investigated whether host factors would explain the relapses. First, susceptibility to amphotericin B of L. infantum isolates was evaluated in vitro. Then, clinical data and the lipid profile of patients with relapsing and non-relapsing VL were assessed. Susceptibility to amphotericin B was similar between the isolates. CD4+ lymphocytes were reduced in both groups of patients in the first episode and with relapsing VL. Still, the strongest blood cell indicator associated with relapses was low total lymphocyte counts. Total plasma cholesterol, high-density lipoprotein, low-density lipoprotein, and, uniquely, triglycerides of the six individuals in the first episode and twenty-three with relapsing VL were lower in relapsing patients than those in the first episode. Deceased patients had extremely low low-density lipoprotein. After CD4+ decreases, lymphocyte CD8+ reduction is the final stage of immunological failure. The lower lipid concentrations appear to be secondary to the depletion of fat stores by inflammation-induced cachexia and fat exhaustion provoked by the co-occurrence of both diseases, which can finally lead to death.
Collapse
Affiliation(s)
- Renata V. S. Silva
- Laboratório de Leishmanioses, Departamento de Medicina Comunitária, Universidade Federal do Piauí, Teresina 64002-510, PI, Brazil;
| | - Silvia R. B. Uliana
- Laboratório de Leishmanioses, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-090, SP, Brazil; (S.R.B.U.); (J.K.U.Y.Y.)
| | - Jenicer K. U. Y. Yasunaka
- Laboratório de Leishmanioses, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-090, SP, Brazil; (S.R.B.U.); (J.K.U.Y.Y.)
| | - Cláudio S. Veloso
- Instituto de Doenças Tropicais “Natan Portella”, Teresina 64002-510, PI, Brazil; (C.S.V.); (D.L.C.)
| | - Emille Sousa
- Laboratório de Leishmanioses, Centro de Inteligência em Agravos Emergentes e Negligenciados, Teresina 64002-510, PI, Brazil; (E.S.); (M.M.L.F.)
| | - Maria M. L. Ferreira
- Laboratório de Leishmanioses, Centro de Inteligência em Agravos Emergentes e Negligenciados, Teresina 64002-510, PI, Brazil; (E.S.); (M.M.L.F.)
| | - Vivianne S. Carvalho
- Centro de Diagnóstico, “Dr. Raul Bacellar”, Fundação Municipal de Saúde, Teresina 64600-000, PI, Brazil;
| | - Gabriel R. Ferreira
- Department of Microbiology-Infectious Disease and Immunology, Faculty of Medicine, University Laval, Quebec, QC G1V 0A6, Canada;
| | - Dorcas L. Costa
- Instituto de Doenças Tropicais “Natan Portella”, Teresina 64002-510, PI, Brazil; (C.S.V.); (D.L.C.)
- Laboratório de Leishmanioses, Centro de Inteligência em Agravos Emergentes e Negligenciados, Teresina 64002-510, PI, Brazil; (E.S.); (M.M.L.F.)
- Departamento de Mãe e Filho, Universidade Federal do Piauí, Teresina 64002-510, PI, Brazil
| | - Carlos H. N. Costa
- Instituto de Doenças Tropicais “Natan Portella”, Teresina 64002-510, PI, Brazil; (C.S.V.); (D.L.C.)
- Laboratório de Leishmanioses, Centro de Inteligência em Agravos Emergentes e Negligenciados, Teresina 64002-510, PI, Brazil; (E.S.); (M.M.L.F.)
- Departamento de Medicina Comunitária, Universidade Federal do Piauí, Teresina 64002-510, PI, Brazil
- Laboratório de Pesquisas em Leishmanioses, Centro de Investigações em Agravos Tropicais Emergentes e Negligenciados, Instituto de Doenças Tropicais “Natan Portella”, Universidade Federal do Piauí, Rua Artur de Vasconcelos 151, Teresina 64002-510, PI, Brazil
| |
Collapse
|
7
|
Costa CHN, Chang KP, Costa DL, Cunha FVM. From Infection to Death: An Overview of the Pathogenesis of Visceral Leishmaniasis. Pathogens 2023; 12:969. [PMID: 37513817 PMCID: PMC10384967 DOI: 10.3390/pathogens12070969] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 07/02/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
Kala-azar, also known as visceral leishmaniasis (VL), is a disease caused by Leishmania infantum and L. donovani. Patients experience symptoms such as fever, weight loss, paleness, and enlarged liver and spleen. The disease also affects immunosuppressed individuals and has an overall mortality rate of up to 10%. This overview explores the literature on the pathogenesis of preclinical and clinical stages, including studies in vitro and in animal models, as well as complications and death. Asymptomatic infection can result in long-lasting immunity. VL develops in a minority of infected individuals when parasites overcome host defenses and multiply in tissues such as the spleen, liver, and bone marrow. Hepatosplenomegaly occurs due to hyperplasia, resulting from parasite proliferation. A systemic inflammation mediated by cytokines develops, triggering acute phase reactants from the liver. These cytokines can reach the brain, causing fever, cachexia and vomiting. Similar to sepsis, disseminated intravascular coagulation (DIC) occurs due to tissue factor overexpression. Anemia, hypergammaglobulinemia, and edema result from the acute phase response. A regulatory response and lymphocyte depletion increase the risk of bacterial superinfections, which, combined with DIC, are thought to cause death. Our understanding of VL's pathogenesis is limited, and further research is needed to elucidate the preclinical events and clinical manifestations in humans.
Collapse
Affiliation(s)
- Carlos H N Costa
- Centro de Investigações em Agravos Tropicais Emergentes e Negligenciados, Instituto de Doenças Tropicais Natan Portella, Universidade Federal do Piauí, Rua Artur de Vasconcelos 151-Sul, Teresina 64002-510, PI, Brazil
| | - Kwang-Poo Chang
- Department of Microbiology/Immunology, Center for Cancer Cell Biology, Immunology & Infection, Chicago Medical School, Rosalind Franklin University, North Chicago, IL 60064, USA
| | - Dorcas L Costa
- Centro de Investigações em Agravos Tropicais Emergentes e Negligenciados, Instituto de Doenças Tropicais Natan Portella, Universidade Federal do Piauí, Rua Artur de Vasconcelos 151-Sul, Teresina 64002-510, PI, Brazil
| | - Francisco Valmor M Cunha
- Departament of Physiotherapy, Centro Universitário Uninovafapi, Rua Vitorino Orthiges Fernandes, 6123-Uruguai, Teresina 64073-505, PI, Brazil
| |
Collapse
|
8
|
Takele Y, Mulaw T, Adem E, Womersley R, Kaforou M, Franssen SU, Levin M, Taylor GP, Müller I, Cotton JA, Kropf P. Recurrent visceral leishmaniasis relapses in HIV co-infected patients are characterized by less efficient immune responses and higher parasite load. iScience 2023; 26:105867. [PMID: 36685039 PMCID: PMC9845767 DOI: 10.1016/j.isci.2022.105867] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/13/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Visceral leishmaniasis (VL) and HIV co-infection (VL/HIV) has emerged as a significant public health problem in Ethiopia, with up to 30% of patients with VL co-infected with HIV. These patients suffer from recurrent VL relapses and increased mortality. Those with a previous history of VL relapses (recurrent VL/HIV) experience increased VL relapses as compared to patients with HIV presenting with their first episode of VL (primary VL/HIV). Our aim was to identify drivers that account for the higher rate of VL relapses in patients with recurrent VL/HIV (n = 28) as compared to primary VL/HIV (n = 21). Our results show that the relapse-free survival in patients with recurrent VL/HIV was shorter, that they had higher parasite load, lower weight gain, and lower recovery of all blood cell lineages. Their poorer prognosis was characterized by lower production of IFN-gamma, lower CD4+ T cell counts, and higher expression of programmed cell death protein 1 (PD1) on T cells.
Collapse
Affiliation(s)
- Yegnasew Takele
- Department of Infectious Disease, Imperial College London, London, UK
- Leishmaniasis Research and Treatment Centre, University of Gondar, Gondar, Ethiopia
| | - Tadele Mulaw
- Leishmaniasis Research and Treatment Centre, University of Gondar, Gondar, Ethiopia
| | - Emebet Adem
- Leishmaniasis Research and Treatment Centre, University of Gondar, Gondar, Ethiopia
| | - Rebecca Womersley
- Department of Infectious Disease, Imperial College London, London, UK
| | - Myrsini Kaforou
- Department of Infectious Disease, Imperial College London, London, UK
| | | | - Michael Levin
- Department of Infectious Disease, Imperial College London, London, UK
| | | | - Ingrid Müller
- Department of Infectious Disease, Imperial College London, London, UK
| | | | - Pascale Kropf
- Department of Infectious Disease, Imperial College London, London, UK
| |
Collapse
|
9
|
Saini I, Joshi J, Kaur S. Unwelcome prevalence of leishmaniasis with several other infectious diseases. Int Immunopharmacol 2022; 110:109059. [DOI: 10.1016/j.intimp.2022.109059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/12/2022] [Accepted: 07/12/2022] [Indexed: 11/17/2022]
|
10
|
Takele Y, Mulaw T, Adem E, Shaw CJ, Franssen SU, Womersley R, Kaforou M, Taylor GP, Levin M, Müller I, Cotton JA, Kropf P. Immunological factors, but not clinical features, predict visceral leishmaniasis relapse in patients co-infected with HIV. Cell Rep Med 2022; 3:100487. [PMID: 35106507 PMCID: PMC8784791 DOI: 10.1016/j.xcrm.2021.100487] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 10/11/2021] [Accepted: 12/07/2021] [Indexed: 11/17/2022]
Abstract
Visceral leishmaniasis (VL) has emerged as a clinically important opportunistic infection in HIV patients, as VL/HIV co-infected patients suffer from frequent VL relapse. Here, we follow cohorts of VL patients with or without HIV in Ethiopia. By the end of the study, 78.1% of VL/HIV-but none of the VL patients-experience VL relapse. Despite a clinically defined cure, VL/HIV patients maintain higher parasite loads, lower BMI, hepatosplenomegaly, and pancytopenia. We identify three immunological markers associated with VL relapse in VL/HIV patients: (1) failure to restore antigen-specific production of IFN-γ, (2) persistently lower CD4+ T cell counts, and (3) higher expression of PD1 on CD4+ and CD8+ T cells. We show that these three markers, which can be measured in primary hospital settings in Ethiopia, combine well in predicting VL relapse. The use of our prediction model has the potential to improve disease management and patient care.
Collapse
Affiliation(s)
- Yegnasew Takele
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
- Leishmaniasis Research and Treatment Centre, University of Gondar, PO Box 196, Gondar, Ethiopia
| | - Tadele Mulaw
- Leishmaniasis Research and Treatment Centre, University of Gondar, PO Box 196, Gondar, Ethiopia
| | - Emebet Adem
- Leishmaniasis Research and Treatment Centre, University of Gondar, PO Box 196, Gondar, Ethiopia
| | - Caroline Jayne Shaw
- Department of Metabolism, Digestion, and Reproduction, Imperial College London, London SW7 2AZ, UK
| | | | - Rebecca Womersley
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
| | - Myrsini Kaforou
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
| | | | - Michael Levin
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
| | - Ingrid Müller
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
| | | | - Pascale Kropf
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
| |
Collapse
|
11
|
Barros-Gonçalves TDD, Saavedra AF, da Silva-Couto L, Ribeiro-Romão RP, Bezerra-Paiva M, Gomes-Silva A, Carvalho VF, Da-Cruz AM, Pinto EF. Increased levels of cortisol are associated with the severity of experimental visceral leishmaniasis in a Leishmania (L.) infantum-hamster model. PLoS Negl Trop Dis 2021; 15:e0009987. [PMID: 34813597 PMCID: PMC8651114 DOI: 10.1371/journal.pntd.0009987] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 12/07/2021] [Accepted: 11/08/2021] [Indexed: 12/03/2022] Open
Abstract
Background Several infectious diseases are associated with hypothalamic-pituitary-adrenal (HPA) axis disorders by elevating circulating glucocorticoids (GCs), which are known to have an immunosuppressive potential. We conducted this study in golden hamsters, a suitable model for human visceral leishmaniasis (VL), to investigate the relationship of Leishmania (L.) infantum infection on cortisol production and VL severity. Methods L. infantum-infected (n = 42) and uninfected hamsters (n = 30) were followed-up at 30, 120, and 180 days post-infection (dpi). Plasma cortisol was analyzed by radioimmunoassay and cytokines, inducible nitric oxide synthase (iNOS), and arginase by RT-qPCR. Results All hamsters showed splenomegaly at 180 dpi. Increased parasite burden was associated with higher arginase expression and lower iNOS induction. Cortisol levels were elevated in infected animals in all-time points evaluated. Except for monocytes, all other leucocytes showed a strong negative correlation with cortisol, while transaminases were positively correlated. Immunological markers as interleukin (IL)-6, IL-1β, IL-10, and transforming growth-factor-β (TGF-β) were positively correlated to cortisol production, while interferon-γ (IFN-γ) presented a negative correlation. A network analysis showed cortisol as an important knot linking clinical status and immunological parameters. Conclusions These results suggest that L. infantum increases the systemic levels of cortisol, which showed to be associated with hematological, biochemical, and immunological parameters associated to VL severity. Visceral leishmaniasis (VL) is an infectious disease that is common in most tropical countries. VL has high morbidity and leads to death if not properly treated. In Brazil, Leishmania (Leishmania) infantum is the main causative agent of VL. Golden hamsters have proven to be a suitable model for VL. Despite the importance of hypothalamic-pituitary-adrenal (HPA) axis disturbances in infectious disease, few studies have addressed this issue in VL. In this study, we showed that L. infantum-infected hamsters present augmented levels of plasmatic cortisol in association with increased spleen parasite burden. Indeed, a strong positive correlation was observed between cortisol and biochemical parameters (AST/ALT/ALP) related to liver damage, as well as pro-inflammatory cytokines (IL-6 and IL-1β), anti-inflammatory cytokines (IL-10 and TGF-β), and the arginase enzyme that may favor the progression of infection. On the other side, cortisol was negatively correlated with leucocytes, except monocytes, and with IFN-γ and iNOS, which are involved in parasite-killing macrophage function. These results shed light on an unexplored aspect of VL pathogenesis, which is the importance of cortisol production in the disease-associated immune dysfunction.
Collapse
Affiliation(s)
| | - Andrea F. Saavedra
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Luzinei da Silva-Couto
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Raquel P. Ribeiro-Romão
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Milla Bezerra-Paiva
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Adriano Gomes-Silva
- Instituto Nacional de Infectologia Evandro Chagas, FIOCRUZ, Rio de Janeiro, Brazil
| | - Vinicius F. Carvalho
- Laboratório de Inflamação, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Neuroimunomodulação (INCT-NIM), CNPq, Rio de Janeiro, Brazil
| | - Alda Maria Da-Cruz
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Neuroimunomodulação (INCT-NIM), CNPq, Rio de Janeiro, Brazil
- Disciplina de Parasitologia-DMIP, Faculdade de Ciências Médicas, UERJ, Rio de Janeiro, Brazil
- Rede de Pesquisas em Saúde do Estado do Rio de Janeiro/FAPERJ, Rio de Janeiro, Brazil
| | - Eduardo F. Pinto
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
- Rede de Pesquisas em Saúde do Estado do Rio de Janeiro/FAPERJ, Rio de Janeiro, Brazil
- * E-mail:
| |
Collapse
|
12
|
Dalzini A, Ballin G, Dominguez-Rodriguez S, Rojo P, Petrara MR, Foster C, Cotugno N, Ruggiero A, Nastouli E, Klein N, Rinaldi S, Pahwa S, Rossi P, Giaquinto C, Palma P, De Rossi A. Size of HIV-1 reservoir is associated with telomere shortening and immunosenescence in early-treated European children with perinatally acquired HIV-1. J Int AIDS Soc 2021; 24:e25847. [PMID: 34797948 PMCID: PMC8604380 DOI: 10.1002/jia2.25847] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 10/28/2021] [Indexed: 12/31/2022] Open
Abstract
Introduction Persistence of HIV‐1, causing chronic immune activation, is a key determinant of premature senescence. Early antiretroviral therapy (ART) has been associated with a reduced HIV‐1 reservoir in children with perinatally acquired HIV‐1 (PHIV), but its impact on the senescence process is an open question. We investigated the association between HIV‐1 reservoir and biological and immune ageing profile in PHIV enrolled in the multicentre cross‐sectional study CARMA (Child and Adolescent Reservoir Measurements on early suppressive ART) conducted within the EPIICAL (Early treated Perinatally HIV Infected individuals: Improving Children's Actual Life) consortium. Methods Between September 2017 and June 2018, CARMA enrolled 40 PHIV who started ART before 2 years of age and had undetectable viremia for at least 5 years before sampling date. Samples from 37 children with a median age of 13.8 years were available for this study. HIV‐1 DNA copies on CD4 cells, relative telomere length (marker of cellular senescence) and levels of T‐cell receptor rearrangement excision circle (TREC, marker of thymic output) on CD4 and CD8 cells were quantified by qPCR. Immunological profile was assessed by flow cytometry. Associations between molecular and phenotypic markers, HIV‐1 reservoir and age at ART initiation were explored using a multivariable Poisson regression. Results Higher HIV‐1 reservoir was associated (p<0.001) with telomere shortening (incidence rate ratio [IRR] = 0.15 [0.13–0.17]), immunosenescence (CD28–CD57+, IRR = 1.23 [1.21–1.26]) and immunoactivation (CD38+ HLADR+, IRR = 7.29 [6.58–8.09]) of CD4 cells. Late ART initiation (after 6 months of age) correlated with higher HIV‐1 reservoir levels (552 [303–1001] vs. 89 [56–365] copies/106 CD4 cells, p = 0.003) and percentage of CD4 senescent cells (2.89 [1.95–6.31] vs. 1.02 [0.45–2.69, p = 0.047). TREC levels in CD8 cells were inversely associated with HIV‐1 reservoir (IRR = 0.77 [0.76–0.79]) and were significantly lower in late treated PHIV (1128 [486–1671] vs. 2278 [1425–3314], p = 0.042). Conclusions Later ART initiation is associated with higher HIV‐1 reservoir size, which correlates with increased telomere shortening and senescence of CD4 cells. Timing of ART initiation in infancy has long‐term consequences on the immune and biological ageing profile of children with perinatally acquired HIV‐1.
Collapse
Affiliation(s)
- Annalisa Dalzini
- Section of Oncology and Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Giovanni Ballin
- Section of Oncology and Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | | | - Pablo Rojo
- Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Maria Raffaella Petrara
- Section of Oncology and Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | | | - Nicola Cotugno
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.,Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Alessandra Ruggiero
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Eleni Nastouli
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Nigel Klein
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Stefano Rinaldi
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Savita Pahwa
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Paolo Rossi
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.,Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Carlo Giaquinto
- Department of Mother and Child Health, University of Padova, Padova, Italy
| | - Paolo Palma
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.,Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Anita De Rossi
- Section of Oncology and Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy.,Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | -
- Section of Oncology and Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| |
Collapse
|
13
|
High levels of anti-Leishmania IgG3 and low CD4 + T cells count were associated with relapses in visceral leishmaniasis. BMC Infect Dis 2021; 21:369. [PMID: 33874901 PMCID: PMC8056614 DOI: 10.1186/s12879-021-06051-5] [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: 12/11/2020] [Accepted: 03/08/2021] [Indexed: 01/10/2023] Open
Abstract
Background Visceral leishmaniasis (VL) is severe and potentially fatal. Brazil is one of the countries with the greatest endemicity for the disease in the world. The reduction of CD4+ T lymphocytes, B cells activation and high levels of inflammatory cytokines (IL-6/IL-8/TNF/IL-1β), plasma LPS, soluble CD14, anti-Leishmania IgG3 and low leptin levels are involved in the immunopathogenesis of VL, most associated with severe VL. Despite relapses occurring in about 4–5% of patients with VL not associated with HIV infection, the factors underlying relapses are little known. Our aim was to identify clinical, laboratory and immunological parameters that may be associated with recurrences in VL. Methods Fifteen VL patients recruited from Hospital Eduardo de Menezes (BH-MG) were grouped into relapsing (R-VL, n = 5) and non-relapsing (NR-VL, n = 10) and evaluated during active disease, immediately after treatment (post-treatment) and 6 months post-treatment (6mpt). Clinical and laboratory data obtained from medical records were correlated with CD4+ and CD8+ T cell counts and anti-Leishmania Igs and IL-6 plasma levels and compared to those parameters of ten healthy controls. Results During the active phase of VL, despite similarity in the clinical symptoms, the rates of thrombocytopenia, elevated transaminases (AST and ALT) and hyperbilirubinemia were higher in the NR-VL group compared to R-VL (p < 0.05), a profile reversed during the post-treatment phase. All patients had low CD4+ T counts in active phase, however, NR-VL patients had a higher gain of this cell type than R-VL in the post-treatment (p < 0.05). There was a significant reduction in IgG3 levels during the follow-up in the NR-VL group compared to the R-VL, especially at 6mpt (p < 0.05). In addition, IgG3 levels were negatively correlated with CD4+ T counts in the R-VL group (r = − 0.52). Elevated levels of IL-6 were observed in active VL and correlated with clinical markers of severity. Conclusions During active phase of VL, the NR-VL patients presented more severe laboratorial abnormalities compared to R-VL, probably because the latter had already received previous treatment. On the other hand, R-VL exhibited greater impairment of immune reconstitution and a high degree of B lymphocyte activation, which must be a factor that favored relapses. Supplementary Information The online version contains supplementary material available at 10.1186/s12879-021-06051-5.
Collapse
|
14
|
Batista MA, Calvo-Fortes F, Silveira-Nunes G, Camatta GC, Speziali E, Turroni S, Teixeira-Carvalho A, Martins-Filho OA, Neretti N, Maioli TU, Santos RR, Brigidi P, Franceschi C, Faria AMC. Inflammaging in Endemic Areas for Infectious Diseases. Front Immunol 2020; 11:579972. [PMID: 33262758 PMCID: PMC7688519 DOI: 10.3389/fimmu.2020.579972] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 10/13/2020] [Indexed: 12/20/2022] Open
Abstract
Immunosenescence is marked by a systemic process named inflammaging along with a series of defects in the immunological activity that results in poor responses to infectious agents and to vaccination. Inflammaging, a state of low-grade chronic inflammation, usually leads to chronic inflammatory diseases and frailty in the elderly. However, some elderly escape from frailty and reach advanced age free of the consequences of inflammaging. This process has been called immunological remodeling, and it is the hallmark of healthy aging as described in the studies of centenarians in Italy. The biological markers of healthy aging are still a matter of debate, and the studies on the topic have focused on inflammatory versus remodeling processes and molecules. The sub-clinical inflammatory status associated with aging might be a deleterious event for populations living in countries where chronic infectious diseases are not prevalent. Nevertheless, in other parts of the world where they are, two possibilities may occur. Inflammatory responses may have a protective effect against these infectious agents. At the same time, the long-term consequences of protective immune responses during chronic infections may result in accelerated immunosenescence in these individuals. Therefore, the biological markers of healthy aging can vary according to environmental, cultural, and geographical settings that reflect worldwide, and in a non-biased, non-westernized perspective, the changes that we experience regarding our contacts with microorganisms and the outcomes of such contacts.
Collapse
Affiliation(s)
- Marina Andrade Batista
- Programa de Pós Graduação em Nutrição e Saúde, Escola de Enfermagem, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fernanda Calvo-Fortes
- Programa de Pós Graduação em Nutrição e Saúde, Escola de Enfermagem, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Gabriela Silveira-Nunes
- Departamento de Medicina, Universidade Federal de Juiz de Fora, Governador Valadares, Brazil
| | - Giovanna Caliman Camatta
- Programa de Pós Graduação em Nutrição e Saúde, Escola de Enfermagem, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Elaine Speziali
- Instituto Rene Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Silvia Turroni
- Unit of Microbial Ecology of Health, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | | | | | - Nicola Neretti
- Departament of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, United States
| | - Tatiani Uceli Maioli
- Programa de Pós Graduação em Nutrição e Saúde, Escola de Enfermagem, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Rodrigo Ribeiro Santos
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Patrizia Brigidi
- Unit of Microbial Ecology of Health, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Claudio Franceschi
- Center for Biophysics, Bioinformatics, Biocomplexity, University of Bologna, Bologna, Italy.,Laboratory of Systems Biology of Healthy Aging, Department of Applied Mathematics, Lobachevsky University, Nizhny Novgorod, Russia
| | - Ana Maria Caetano Faria
- Programa de Pós Graduação em Nutrição e Saúde, Escola de Enfermagem, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|