1
|
Buthasane W, Shotelersuk V, Chetruengchai W, Srichomthong C, Assawapitaksakul A, Tangphatsornruang S, Pootakham W, Sonthirod C, Tongsima S, Wangkumhang P, Wilantho A, Thongphakdee A, Sanannu S, Poksawat C, Nipanunt T, Kasorndorkbua C, Koepfli KP, Pukazhenthi BS, Suriyaphol P, Wongsurawat T, Jenjaroenpun P, Suriyaphol G. Comprehensive genome assembly reveals genetic diversity and carcass consumption insights in critically endangered Asian king vultures. Sci Rep 2024; 14:9455. [PMID: 38658744 PMCID: PMC11043450 DOI: 10.1038/s41598-024-59990-9] [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: 11/23/2023] [Accepted: 04/17/2024] [Indexed: 04/26/2024] Open
Abstract
The Asian king vulture (AKV), a vital forest scavenger, is facing globally critical endangerment. This study aimed to construct a reference genome to unveil the mechanisms underlying its scavenger abilities and to assess the genetic relatedness of the captive population in Thailand. A reference genome of a female AKV was assembled from sequencing reads obtained from both PacBio long-read and MGI short-read sequencing platforms. Comparative genomics with New World vultures (NWVs) and other birds in the Family Accipitridae revealed unique gene families in AKV associated with retroviral genome integration and feather keratin, contrasting with NWVs' genes related to olfactory reception. Expanded gene families in AKV were linked to inflammatory response, iron regulation and spermatogenesis. Positively selected genes included those associated with anti-apoptosis, immune response and muscle cell development, shedding light on adaptations for carcass consumption and high-altitude soaring. Using restriction site-associated DNA sequencing (RADseq)-based genome-wide single nucleotide polymorphisms (SNPs), genetic relatedness and inbreeding status of five captive AKVs were determined, revealing high genomic inbreeding in two females. In conclusion, the AKV reference genome was established, providing insights into its unique characteristics. Additionally, the potential of RADseq-based genome-wide SNPs for selecting AKV breeders was demonstrated.
Collapse
Affiliation(s)
- Wannapol Buthasane
- Biochemistry Unit, Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Vorasuk Shotelersuk
- Center of Excellence for Medical Genomics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Henri Dunant Road, Pathumwan, Bangkok, 10330, Thailand
- Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, 10330, Thailand
| | - Wanna Chetruengchai
- Center of Excellence for Medical Genomics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Henri Dunant Road, Pathumwan, Bangkok, 10330, Thailand
- Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, 10330, Thailand
| | - Chalurmpon Srichomthong
- Center of Excellence for Medical Genomics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Henri Dunant Road, Pathumwan, Bangkok, 10330, Thailand
- Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, 10330, Thailand
| | - Adjima Assawapitaksakul
- Center of Excellence for Medical Genomics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Henri Dunant Road, Pathumwan, Bangkok, 10330, Thailand
- Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, 10330, Thailand
| | - Sithichoke Tangphatsornruang
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani, 12120, Thailand
| | - Wirulda Pootakham
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani, 12120, Thailand
| | - Chutima Sonthirod
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani, 12120, Thailand
| | - Sissades Tongsima
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani, 12120, Thailand
| | - Pongsakorn Wangkumhang
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani, 12120, Thailand
| | - Alisa Wilantho
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani, 12120, Thailand
| | - Ampika Thongphakdee
- Animal Conservation and Research Institute, The Zoological Park Organization of Thailand under the Royal Patronage of H.M. The King, Bangkok, 10300, Thailand
| | - Saowaphang Sanannu
- Animal Conservation and Research Institute, The Zoological Park Organization of Thailand under the Royal Patronage of H.M. The King, Bangkok, 10300, Thailand
| | - Chaianan Poksawat
- Animal Conservation and Research Institute, The Zoological Park Organization of Thailand under the Royal Patronage of H.M. The King, Bangkok, 10300, Thailand
| | - Tarasak Nipanunt
- Huai Kha Khaeng Wildlife Breeding Center, Department of National Parks, Wildlife and Plant Conservation, Uthai Thani, 61160, Thailand
| | - Chaiyan Kasorndorkbua
- Laboratory of Raptor Research and Conservation Medicine, Department of Pathology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, 10900, Thailand
| | - Klaus-Peter Koepfli
- Smithsonian-Mason School of Conservation, George Mason University, Front Royal, VA, 22630, USA
- Center for Species Survival, Smithsonian Conservation Biology Institute, National Zoological Park, Front Royal, VA, 22630, USA
| | - Budhan S Pukazhenthi
- Center for Species Survival, Smithsonian Conservation Biology Institute, National Zoological Park, Front Royal, VA, 22630, USA
| | - Prapat Suriyaphol
- Division of Medical Bioinformatics, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Thidathip Wongsurawat
- Division of Medical Bioinformatics, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Piroon Jenjaroenpun
- Division of Medical Bioinformatics, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Gunnaporn Suriyaphol
- Biochemistry Unit, Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
2
|
Stanton DJ, Quadri NZ, Tanabe MB. Concomitantly Diagnosed Disseminated M kansasii Infection and Hairy Cell Leukemia With Review of Pathophysiology. J Investig Med High Impact Case Rep 2024; 12:23247096241253343. [PMID: 38767131 PMCID: PMC11107317 DOI: 10.1177/23247096241253343] [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: 03/17/2024] [Revised: 04/08/2024] [Accepted: 04/21/2024] [Indexed: 05/22/2024] Open
Abstract
The association between Hairy Cell Leukemia (HCL) and non-tuberculous mycobacterial infections (NTMs) is well described, most notably Mycobacterium kansasii. The exact pathophysiology is not known. We report a case of a 31-year-old male with concomitantly diagnosed HCL and disseminated M kansasii infection who presented with rash, pancytopenia, and bulky axillary lymphadenopathy. The M kansasii was initially diagnosed through use of cell-free DNA detection and confirmed by bone marrow and lymph node cultures. Hairy Cell Leukemia was diagnosed with peripheral flow cytometry and confirmed via the same bone marrow sample. His HCL was put into remission with a single course of cladribine and rituximab chemotherapy; however, his M kansasii infection persisted for 6 months despite aggressive antimicrobial and surgical therapy. It was finally controlled using high-dose rifampin in combination with azithromycin and ethambutol. This case highlights the known link between HCL and M kansasii. Furthermore, it hints at potential causes beyond chemotherapy-induced immunocompromise. Notable possibilities include HCL cells acting as sanctuary sites for M kansasii to evade the immune system, and subclinical M kansasii infections causing NLRP3 inflammasome overactivation to trigger the oncogenic transformation to HCL. More research into the pathophysiologic link between HCL and M kansasii infections would allow for more effective prevention, diagnosis, and treatment of these severe atypical infections which are the major cause of morbidity in the cladribine era of HCL treatment.
Collapse
|
3
|
Teichman J, Geddes M, Zhu N, Keating MM, Sabloff M, Christou G, Leber B, Khalaf D, St-Hilaire E, Finn N, Shamy A, Yee KW, Storring JM, Nevill TJ, Delage R, Elemary M, Banerji V, Houston B, Mozessohn L, Chodirker L, Zhang L, Siddiqui M, Parmentier A, Leitch HA, Buckstein RJ. High transferrin saturation predicts inferior clinical outcomes in patients with myelodysplastic syndromes. Haematologica 2022; 108:532-542. [PMID: 35979720 PMCID: PMC9890030 DOI: 10.3324/haematol.2022.280723] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Indexed: 02/03/2023] Open
Abstract
Iron overload (IO) reflected by elevated ferritin is associated with increased mortality in myelodysplastic syndromes (MDS), however, ferritin is an imperfect metric. Elevated labile plasma iron correlates with clinical outcomes and transferrin saturation (TSAT) >80%, but is not readily measurable. The trajectory of TSAT, and its association with clinical outcomes remain undefined. Canadian MDS registry patients were evaluated. Mean TSAT, mean ferritin and transfusion dose density (TDD) were determined. Survival was evaluated by TSAT and ferritin (<50%, 50-80%, >80%), (≤500 μg/L, 501-800 μg/L, >800 μg/L). In 718 patients, median age was 74 years; 12%, 31%, 29%, 15% and 13% were IPSS-R very low, low, intermediate, high and very high. TSAT and ferritin were moderately correlated (r=0.63, P<0.0001). TSAT increased over time in transfusion- dependent patients (P=0.006). Higher TSAT and ferritin were associated with inferior 5-year overall (OS), progression- free (PFS), and leukemia-free survival (LFS) (P≤0.008) and higher TDD with inferior 5-year OS. TSAT >80% trended with inferior cardiac death-free survival (P=0.053). In univariate analysis, age, IPSS-R, blast percentage by Eastern Cooperative Oncology Group Performance Status, frailty, Charlson Comorbidity Index, iron chelation (Y/N), TDD, TSAT and ferritin were significantly associated with inferior OS. By multivariable analysis, TSAT >80% (P=0.007) remained significant for OS (R2 30.3%). In MDS, TSAT >80% and ferritin >800 μg/L portended inferior OS, PFS and LFS. TSAT may indicate the presence of oxidative stress, and is readily measurable in a clinical setting. The relationship between TSAT and cardiac death-free survival warrants further study.
Collapse
Affiliation(s)
| | | | - Nancy Zhu
- University of Alberta, Edmonton, Alberta
| | | | | | | | | | | | - Eve St-Hilaire
- Dr. Georges-L-Dumont University Hospital Center, Moncton, New Brunswick
| | - Nicholas Finn
- Dr. Georges-L-Dumont University Hospital Center, Moncton, New Brunswick
| | | | | | | | | | | | | | - Versha Banerji
- Sunnybrook Health Sciences Center, Toronto, Ontario,QEII Health Sciences Centre, Halifax, Nova Scotia
| | | | | | | | - Liying Zhang
- Sunnybrook Health Sciences Center, Toronto, Ontario
| | | | | | - Heather A. Leitch
- St. Paul’s Hospital, Vancouver, British Columbia, Canada,HAL and RJB contributed equally as co-senior authors
| | - Rena J. Buckstein
- Sunnybrook Health Sciences Center, Toronto, Ontario,HAL and RJB contributed equally as co-senior authors
| |
Collapse
|
4
|
Moreira AC, Silva T, Mesquita G, Gomes AC, Bento CM, Neves JV, Rodrigues DF, Rodrigues PN, Almeida AA, Santambrogio P, Gomes MS. H-Ferritin Produced by Myeloid Cells Is Released to the Circulation and Plays a Major Role in Liver Iron Distribution during Infection. Int J Mol Sci 2021; 23:ijms23010269. [PMID: 35008695 PMCID: PMC8745395 DOI: 10.3390/ijms23010269] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/22/2021] [Accepted: 12/24/2021] [Indexed: 11/16/2022] Open
Abstract
During infections, the host redistributes iron in order to starve pathogens from this nutrient. Several proteins are involved in iron absorption, transport, and storage. Ferritin is the most important iron storage protein. It is composed of variable proportions of two peptides, the L- and H-ferritins (FTL and FTH). We previously showed that macrophages increase their expression of FTH1 when they are infected in vitro with Mycobacterium avium, without a significant increase in FTL. In this work, we investigated the role of macrophage FTH1 in M. avium infection in vivo. We found that mice deficient in FTH1 in myeloid cells are more resistant to M. avium infection, presenting lower bacterial loads and lower levels of proinflammatory cytokines than wild-type littermates, due to the lower levels of available iron in the tissues. Importantly, we also found that FTH1 produced by myeloid cells in response to infection may be found in circulation and that it plays a key role in iron redistribution. Specifically, in the absence of FTH1 in myeloid cells, increased expression of ferroportin is observed in liver granulomas and increased iron accumulation occurs in hepatocytes. These results highlight the importance of FTH1 expression in myeloid cells for iron redistribution during infection.
Collapse
Affiliation(s)
- Ana C. Moreira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (A.C.M.); (T.S.); (A.C.G.); (C.M.B.); (J.V.N.); (D.F.R.); (P.N.R.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal;
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Tânia Silva
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (A.C.M.); (T.S.); (A.C.G.); (C.M.B.); (J.V.N.); (D.F.R.); (P.N.R.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal;
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Gonçalo Mesquita
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal;
| | - Ana Cordeiro Gomes
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (A.C.M.); (T.S.); (A.C.G.); (C.M.B.); (J.V.N.); (D.F.R.); (P.N.R.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal;
| | - Clara M. Bento
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (A.C.M.); (T.S.); (A.C.G.); (C.M.B.); (J.V.N.); (D.F.R.); (P.N.R.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal;
- Programa Doutoral em Biologia Molecular e Celular (MCbiology), Instituto de Ciências Biomédicas Abel Salazar da Universidade do Porto, 4200-135 Porto, Portugal
| | - João V. Neves
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (A.C.M.); (T.S.); (A.C.G.); (C.M.B.); (J.V.N.); (D.F.R.); (P.N.R.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal;
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Daniela F. Rodrigues
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (A.C.M.); (T.S.); (A.C.G.); (C.M.B.); (J.V.N.); (D.F.R.); (P.N.R.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal;
| | - Pedro N. Rodrigues
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (A.C.M.); (T.S.); (A.C.G.); (C.M.B.); (J.V.N.); (D.F.R.); (P.N.R.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal;
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Agostinho A. Almeida
- LAQV/REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, 4050-313 Porto, Portugal;
| | - Paolo Santambrogio
- Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy;
| | - Maria Salomé Gomes
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (A.C.M.); (T.S.); (A.C.G.); (C.M.B.); (J.V.N.); (D.F.R.); (P.N.R.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal;
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
- Correspondence:
| |
Collapse
|
5
|
Moreira AC, Teles MJ, Silva T, Bento CM, Alves IS, Pereira L, Guimarães JT, Porto G, Oliveira P, Gomes MS. Iron Related Biomarkers Predict Disease Severity in a Cohort of Portuguese Adult Patients during COVID-19 Acute Infection. Viruses 2021; 13:v13122482. [PMID: 34960751 PMCID: PMC8703662 DOI: 10.3390/v13122482] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/03/2021] [Accepted: 12/07/2021] [Indexed: 02/07/2023] Open
Abstract
Large variability in COVID-19 clinical progression urges the need to find the most relevant biomarkers to predict patients' outcomes. We evaluated iron metabolism and immune response in 303 patients admitted to the main hospital of the northern region of Portugal with variable clinical pictures, from September to November 2020. One hundred and twenty-seven tested positive for SARS-CoV-2 and 176 tested negative. Iron-related laboratory parameters and cytokines were determined in blood samples collected soon after admission. Demographic data, comorbidities and clinical outcomes were recorded. Patients were assigned into five groups according to severity. Serum iron and transferrin levels at admission were lower in COVID-19-positive than in COVID-19-negative patients. The levels of interleukin (IL)-6 and monocyte chemoattractant protein 1 (MCP-1) were increased in COVID-19-positive patients. The lowest serum iron and transferrin levels at diagnosis were associated with the worst outcomes. Iron levels negatively correlated with IL-6 and higher levels of this cytokine were associated with a worse prognosis. Serum ferritin levels at diagnosis were higher in COVID-19-positive than in COVID-19-negative patients. Serum iron is the simplest laboratory test to be implemented as a predictor of disease progression in COVID-19-positive patients.
Collapse
Affiliation(s)
- Ana C. Moreira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (A.C.M.); (M.J.T.); (T.S.); (C.M.B.); (I.S.A.); (L.P.); (G.P.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal;
| | - Maria Jose Teles
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (A.C.M.); (M.J.T.); (T.S.); (C.M.B.); (I.S.A.); (L.P.); (G.P.)
- CHUSJ—Centro Hospitalar Universitário São João, 4200-319 Porto, Portugal;
- ISPUP-EPIUnit—Instituto de Saúde Pública da Universidade do Porto, 4050-091 Porto, Portugal
| | - Tânia Silva
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (A.C.M.); (M.J.T.); (T.S.); (C.M.B.); (I.S.A.); (L.P.); (G.P.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal;
| | - Clara M. Bento
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (A.C.M.); (M.J.T.); (T.S.); (C.M.B.); (I.S.A.); (L.P.); (G.P.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- MCBiology—Programa Doutoral em Biologia Molecular e Celular, Instituto de Ciências Biomédicas Abel Salazar da Universidade do Porto, 4200-135 Porto, Portugal
| | - Inês Simões Alves
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (A.C.M.); (M.J.T.); (T.S.); (C.M.B.); (I.S.A.); (L.P.); (G.P.)
| | - Luisa Pereira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (A.C.M.); (M.J.T.); (T.S.); (C.M.B.); (I.S.A.); (L.P.); (G.P.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular da Universidade do Porto, 4200-135 Porto, Portugal
| | - João Tiago Guimarães
- CHUSJ—Centro Hospitalar Universitário São João, 4200-319 Porto, Portugal;
- ISPUP-EPIUnit—Instituto de Saúde Pública da Universidade do Porto, 4050-091 Porto, Portugal
- FMUP—Faculdade de Medicina da Universidade do Porto, 4200-319 Porto, Portugal
| | - Graça Porto
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (A.C.M.); (M.J.T.); (T.S.); (C.M.B.); (I.S.A.); (L.P.); (G.P.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal;
- CHPorto—Centro Hospitalar do Porto, 4099-001 Porto, Portugal
| | - Pedro Oliveira
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal;
- ISPUP-EPIUnit—Instituto de Saúde Pública da Universidade do Porto, 4050-091 Porto, Portugal
| | - Maria Salomé Gomes
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (A.C.M.); (M.J.T.); (T.S.); (C.M.B.); (I.S.A.); (L.P.); (G.P.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal;
- Correspondence:
| |
Collapse
|
6
|
Nairz M, Metzendorf C, Vujic-Spasic M, Mitterstiller AM, Schroll A, Haschka D, Hoffmann A, Von Raffay L, Sparla R, Huck CW, Talasz H, Moser PL, Muckenthaler MU, Weiss G. Cell-specific expression of Hfe determines the outcome of Salmonella enterica serovar Typhimurium infection in mice. Haematologica 2021; 106:3149-3161. [PMID: 33054105 PMCID: PMC8634192 DOI: 10.3324/haematol.2019.241745] [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: 12/03/2019] [Accepted: 09/30/2020] [Indexed: 12/05/2022] Open
Abstract
Mutations in HFE cause hereditary hemochromatosis type I hallmarked by increased iron absorption, iron accumulation in hepatocytes and iron deficiency in myeloid cells. HFE encodes an MHC-I like molecule, but its function in immune responses to infection remains incompletely understood. Here, we investigated putative roles of Hfe in myeloid cells and hepatocytes, separately, upon infection with Salmonella Typhimurium, an intracellular bacterium with iron-dependent virulence. We found that constitutive and macrophage-specific deletion of Hfe protected infected mice. The propagation of Salmonella in macrophages was reduced due to limited intramacrophage iron availability for bacterial growth and increased expression of the anti-microbial enzyme nitric oxide synthase-2. By contrast, mice with hepatocyte-specific deletion of Hfe succumbed earlier to Salmonella infection because of unrestricted extracellular bacterial replication associated with high iron availability in the serum and impaired expression of essential host defense molecules such as interleukin-6, interferon-γ and nitric oxide synthase-2. Wild-type mice subjected to dietary iron overload phenocopied hepatocyte-specific Hfe deficiency suggesting that increased iron availability in the serum is deleterious in Salmonella infection and underlies impaired host immune responses. Moreover, the macrophage-specific effect is dominant over hepatocyte-specific Hfe-depletion, as Hfe knock-out mice have increased survival despite the higher parenchymal iron load associated with systemic loss of Hfe. We conclude that cell-specific expression of Hfe in hepatocytes and macrophages differentially affects the course of infections with specific pathogens by determining bacterial iron access and the efficacy of anti-microbial immune effector pathways. This may explain the high frequency and evolutionary conservation of human HFE mutations.
Collapse
Affiliation(s)
- Manfred Nairz
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, 6020 Innsbruck
| | - Christoph Metzendorf
- Department of Pediatric Hematology, Oncology and Immunology, University of Heidelberg, INF 350, 69120 Heidelberg, Germany; Molecular Medicine Partnership Unit, 69120 Heidelberg
| | - Maja Vujic-Spasic
- Department of Pediatric Hematology, Oncology and Immunology, University of Heidelberg, INF 350, 69120 Heidelberg, Germany; Molecular Medicine Partnership Unit, 69120 Heidelberg, Germany; Institute of Comparative Molecular Endocrinology, Ulm University, 89081 Ulm
| | - Anna-Maria Mitterstiller
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, 6020 Innsbruck
| | - Andrea Schroll
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, 6020 Innsbruck
| | - David Haschka
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, 6020 Innsbruck
| | - Alexander Hoffmann
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, 6020 Innsbruck, Austria; Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, 6020 Innsbruck
| | - Laura Von Raffay
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, 6020 Innsbruck
| | - Richard Sparla
- Department of Pediatric Hematology, Oncology and Immunology, University of Heidelberg, INF 350, 69120 Heidelberg, Germany; Molecular Medicine Partnership Unit, 69120 Heidelberg
| | - Christian W Huck
- Institute for Analytical Chemistry and Radiochemistry, University of Innsbruck, 6020 Innsbruck
| | - Heribert Talasz
- Biocenter, Division of Clinical Biochemistry, Medical University of Innsbruck, 6020 Innsbruck
| | | | - Martina U Muckenthaler
- Department of Pediatric Hematology, Oncology and Immunology, University of Heidelberg, INF 350, 69120 Heidelberg, Germany; Molecular Medicine Partnership Unit, 69120 Heidelberg.
| | - Günter Weiss
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, 6020 Innsbruck, Austria; Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, 6020 Innsbruck.
| |
Collapse
|
7
|
Jha V, Pal R, Kumar D, Mukhopadhyay S. ESAT-6 Protein of Mycobacterium tuberculosis Increases Holotransferrin-Mediated Iron Uptake in Macrophages by Downregulating Surface Hemochromatosis Protein HFE. THE JOURNAL OF IMMUNOLOGY 2020; 205:3095-3106. [PMID: 33148716 DOI: 10.4049/jimmunol.1801357] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 10/01/2020] [Indexed: 11/19/2022]
Abstract
Iron is an essential element for Mycobacterium tuberculosis; it has at least 40 enzymes that require iron as a cofactor. Accessibility of iron at the phagosomal surface inside macrophage is crucial for survival and virulence of M. tuberculosis ESAT-6, a 6-kDa-secreted protein of region of difference 1, is known to play a crucial role in virulence and pathogenesis of M. tuberculosis In our earlier study, we demonstrated that ESAT-6 protein interacts with β-2-microglobulin (β2M) and affects class I Ag presentation through sequestration of β2M inside endoplasmic reticulum, which contributes toward inhibition of MHC class I:β2M:peptide complex formation. The 6 aa at C-terminal region of ESAT-6 are essential for ESAT6:β2M interaction. β2M is essential for proper folding of HFE, CD1, and MHC class I and their surface expression. It is known that M. tuberculosis recruit holotransferrin at the surface of the phagosome. But the upstream mechanism by which it modulates holotransferrin-mediated iron uptake at the surface of macrophage is not well understood. In the current study, we report that interaction of the ESAT-6 protein with β2M causes downregulation of surface HFE, a protein regulating iron homeostasis via interacting with transferrin receptor 1 (TFR1). We found that ESAT-6:β2M interaction leads to sequestration of HFE in endoplasmic reticulum, causing poorer surface expression of HFE and HFE:TFR1 complex (nonfunctional TFR1) in peritoneal macrophages from C57BL/6 mice, resulting in increased holotransferrin-mediated iron uptake in these macrophages. These studies suggest that M. tuberculosis probably targets the ESAT-6 protein to increase iron uptake.
Collapse
Affiliation(s)
- Vishwanath Jha
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics, Uppal, Hyderabad 500039, Telangana, India.,Graduate Studies, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India; and
| | - Ravi Pal
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics, Uppal, Hyderabad 500039, Telangana, India.,Graduate Studies, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India; and
| | - Dhiraj Kumar
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
| | - Sangita Mukhopadhyay
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics, Uppal, Hyderabad 500039, Telangana, India;
| |
Collapse
|
8
|
Rauf A, Shariati MA, Khalil AA, Bawazeer S, Heydari M, Plygun S, Laishevtcev A, Hussain MB, Alhumaydhi FA, Aljohani ASM. Hepcidin, an overview of biochemical and clinical properties. Steroids 2020; 160:108661. [PMID: 32450084 DOI: 10.1016/j.steroids.2020.108661] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/31/2020] [Accepted: 05/17/2020] [Indexed: 12/16/2022]
Abstract
Hepcidin is a peptide hormone which helps in regulating iron homeostasis in the human body. Iron obtained from daily diet is passed through the intestinal enterocyte apical membrane via divalent metal transporter 1 (DMT1), which is either stored as ferritin or moved into the plasma by hepcidin-ferroportin (Fpn) as an exporter. Hepcidin (hepatic bactericidal protein) is a cysteine rich peptide, was initially identified as a urinary antimicrobial peptide. It contains 25 amino acids and four disulfide bridges. It has significant role in regulation of iron in the body. Stimulation of iron in plasma and further its storage is linked with the production of hepcidin. This enhancement of iron hampers the absorption of iron from the diet. The cause of hereditary recessive anemia also known as Iron-refractory iron deficiency anemia (IRIDA) is characterized by increased hepcidin production due to a gene mutation in the suppressor matriptase-2/TMPRSS6. During infection, hepcidin plays a defensive role against various infections by depleting the extracellular iron from the body. Moreover, hepcidin lowers the concentrations of iron from the duodenal enterocytes, macrophages and also decrease its transport across the placenta.This review highlights the significant role of hepcidin in the iron homeostasis and as an antimicrobial agent.
Collapse
Affiliation(s)
- Abdur Rauf
- Department of Chemistry, University of Swabi, Anbar 23561, Khyber Pakhtunkhwa, Pakistan.
| | - Mohammad Ali Shariati
- Laboratory of Biocontrol and Antimicrobial Resistance, Orel State University Named After I.S. Turgenev, 302026 Orel, Russia
| | - Anees Ahmed Khalil
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health Sciences, The University of Lahore, Pakistan
| | - Saud Bawazeer
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Umm Al-Qura University, Makkah, P.O. Box 42, Saudi Arabia
| | - Mojtaba Heydari
- Poostchi Ophthalmology Research Center, Shiraz University of Medical Sciences, Shiraz, Sciences, Shiraz, Iran
| | - Sergey Plygun
- Laboratory of Biocontrol and Antimicrobial Resistance, Orel State University Named After I.S. Turgenev, 302026 Orel, Russia; European Society of Clinical Microbiology and Infectious Diseases, Basel 4051, Switzerland; Russian Research Institute of Phytopathology, Moscow Region 143050, Russia
| | - Alexy Laishevtcev
- Laboratory of Biocontrol and Antimicrobial Resistance, Orel State University Named After I.S. Turgenev, 302026 Orel, Russia; Federal Research Center - All-Russian Scientific Research Institute of Experimental Veterinary Medicine named after K.I. Skryabin and Y.R. Kovalenko of the Russian Academy of Sciences, Moscow 109428, Russia
| | - Muhammad Bilal Hussain
- Institute of Home and Food Sciences, Government College University, Faisalabad, Pakistan
| | - Fahad A Alhumaydhi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Abdullah S M Aljohani
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, Qassim University, Buraydah, Saudi Arabia
| |
Collapse
|
9
|
Moreira AC, Mesquita G, Gomes MS. Ferritin: An Inflammatory Player Keeping Iron at the Core of Pathogen-Host Interactions. Microorganisms 2020; 8:microorganisms8040589. [PMID: 32325688 PMCID: PMC7232436 DOI: 10.3390/microorganisms8040589] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 04/10/2020] [Accepted: 04/15/2020] [Indexed: 12/14/2022] Open
Abstract
Iron is an essential element for virtually all cell types due to its role in energy metabolism, nucleic acid synthesis and cell proliferation. Nevertheless, if free, iron induces cellular and organ damage through the formation of free radicals. Thus, iron levels must be firmly controlled. During infection, both host and microbe need to access iron and avoid its toxicity. Alterations in serum and cellular iron have been reported as important markers of pathology. In this regard, ferritin, first discovered as an iron storage protein, has emerged as a biomarker not only in iron-related disorders but also in inflammatory diseases, or diseases in which inflammation has a central role such as cancer, neurodegeneration or infection. The basic research on ferritin identification and functions, as well as its role in diseases with an inflammatory component and its potential as a target in host-directed therapies, are the main considerations of this review.
Collapse
Affiliation(s)
- Ana C. Moreira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (G.M.); (M.S.G.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
- Correspondence:
| | - Gonçalo Mesquita
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (G.M.); (M.S.G.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Maria Salomé Gomes
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (G.M.); (M.S.G.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| |
Collapse
|
10
|
Gomes AC, Moreira AC, Silva T, Neves JV, Mesquita G, Almeida AA, Barreira-Silva P, Fernandes R, Resende M, Appelberg R, Rodrigues PNS, Gomes MS. IFN-γ–Dependent Reduction of Erythrocyte Life Span Leads to Anemia during Mycobacterial Infection. THE JOURNAL OF IMMUNOLOGY 2019; 203:2485-2496. [DOI: 10.4049/jimmunol.1900382] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 09/04/2019] [Indexed: 12/26/2022]
|
11
|
González-Ruiz S, Strillacci MG, Durán-Aguilar M, Cantó-Alarcón GJ, Herrera-Rodríguez SE, Bagnato A, Guzmán LF, Milián-Suazo F, Román-Ponce SI. Genome-Wide Association Study in Mexican Holstein Cattle Reveals Novel Quantitative Trait Loci Regions and Confirms Mapped Loci for Resistance to Bovine Tuberculosis. Animals (Basel) 2019; 9:E636. [PMID: 31480266 PMCID: PMC6769677 DOI: 10.3390/ani9090636] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 08/23/2019] [Accepted: 08/24/2019] [Indexed: 12/26/2022] Open
Abstract
Bovine tuberculosis (bTB) is a disease of cattle that represents a risk to public health and causes severe economic losses to the livestock industry. Recently, genetic studies, like genome-wide association studies (GWAS) have greatly improved the investigation of complex diseases identifying thousands of disease-associated genomic variants. Here, we present evidence of genetic variants associated with resistance to TB in Mexican dairy cattle using a case-control approach with a selective DNA pooling experimental design. A total of 154 QTLRs (quantitative trait loci regions) at 10% PFP (proportion of false positives), 42 at 5% PFP and 5 at 1% PFP have been identified, which harbored 172 annotated genes. On BTA13, five new QTLRs were identified in the MACROD2 and KIF16B genes, supporting their involvement in resistance to bTB. Six QTLRs harbor seven annotated genes that have been previously reported as involved in immune response against Mycobacterium spp: BTA (Bos taurus autosome) 1 (CD80), BTA3 (CTSS), BTA 3 (FCGR1A), BTA 23 (HFE), BTA 25 (IL21R), and BTA 29 (ANO9 and SIGIRR). We identified novel QTLRs harboring genes involved in Mycobacterium spp. immune response. This is a first screening for resistance to TB infection on Mexican dairy cattle based on a dense SNP (Single Nucleotide Polymorphism) chip.
Collapse
Affiliation(s)
- Sara González-Ruiz
- Doctorado en Ciencias Biológicas, Universidad Autónoma de Querétaro, Avenida de las Ciencias S/N Juriquilla, Delegación Santa Rosa Jáuregui, Querétaro C.P. 76230, Mexico
| | - Maria G Strillacci
- Department of Veterinary Medicine, Università degli Studi di Milano, Via Trentacoste, 2, 20134 Milano, Italy.
| | - Marina Durán-Aguilar
- Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Avenida de las Ciencias S/N Juriquilla, Delegación Santa Rosa Jáuregui, Querétaro C.P. 76230, Mexico
| | - Germinal J Cantó-Alarcón
- Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Avenida de las Ciencias S/N Juriquilla, Delegación Santa Rosa Jáuregui, Querétaro C.P. 76230, Mexico
| | - Sara E Herrera-Rodríguez
- Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco A.C., Guadalajara C.P. 44270, Mexico
| | - Alessandro Bagnato
- Department of Veterinary Medicine, Università degli Studi di Milano, Via Trentacoste, 2, 20134 Milano, Italy
| | - Luis F Guzmán
- Centro Nacional de Recursos Genéticos, INIFAP, Tepatitlán de Morelos 47600, Mexico
| | - Feliciano Milián-Suazo
- Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Avenida de las Ciencias S/N Juriquilla, Delegación Santa Rosa Jáuregui, Querétaro C.P. 76230, Mexico
| | - Sergio I Román-Ponce
- Centro Nacional de Investigación Disciplinaria en Fisiología y Mejoramiento animal, INIFAP, SAGARPA, Km. 1 Carretera a Colón, Ajuchitlán, Colón, Querétaro C.P. 76280, Mexico.
| |
Collapse
|
12
|
Iron Supplementation Therapy, A Friend and Foe of Mycobacterial Infections? Pharmaceuticals (Basel) 2019; 12:ph12020075. [PMID: 31108902 PMCID: PMC6630247 DOI: 10.3390/ph12020075] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 05/14/2019] [Accepted: 05/15/2019] [Indexed: 12/21/2022] Open
Abstract
Iron is an essential element that is required for oxygen transfer, redox, and metabolic activities in mammals and bacteria. Mycobacteria, some of the most prevalent infectious agents in the world, require iron as growth factor. Mycobacterial-infected hosts set up a series of defense mechanisms, including systemic iron restriction and cellular iron distribution, whereas mycobacteria have developed sophisticated strategies to acquire iron from their hosts and to protect themselves from iron’s harmful effects. Therefore, it is assumed that host iron and iron-binding proteins, and natural or synthetic chelators would be keys targets to inhibit mycobacterial proliferation and may have a therapeutic potential. Beyond this hypothesis, recent evidence indicates a host protective effect of iron against mycobacterial infections likely through promoting remodeled immune response. In this review, we discuss experimental procedures and clinical observations that highlight the role of the immune response against mycobacteria under various iron availability conditions. In addition, we discuss the clinical relevance of our knowledge regarding host susceptibility to mycobacteria in the context of iron availability and suggest future directions for research on the relationship between host iron and the immune response and the use of iron as a therapeutic agent.
Collapse
|
13
|
Tuning the Anti(myco)bacterial Activity of 3-Hydroxy-4-pyridinone Chelators through Fluorophores. Pharmaceuticals (Basel) 2018; 11:ph11040110. [PMID: 30347802 PMCID: PMC6316862 DOI: 10.3390/ph11040110] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 10/17/2018] [Accepted: 10/18/2018] [Indexed: 02/08/2023] Open
Abstract
Controlling the sources of Fe available to pathogens is one of the possible strategies that can be successfully used by novel antibacterial drugs. We focused our interest on the design of chelators to address Mycobacterium avium infections. Taking into account the molecular structure of mycobacterial siderophores and considering that new chelators must be able to compete for Fe(III), we selected ligands of the 3-hydroxy-4-pyridinone class to achieve our purpose. After choosing the type of chelating unit it was also our objective to design chelators that could be monitored inside the cell and for that reason we designed chelators that could be functionalized with fluorophores. We didn’t realize at the time that the incorporation a fluorophore, to allow spectroscopic detection, would be so relevant for the antimycobacterial effect or to determine the affinity of the chelators towards biological membranes. From a biophysical perspective, this is a fascinating illustration of the fact that functionalization of a molecule with a particular label may lead to a change in its membrane permeation properties and result in a dramatic change in biological activity. For that reason we believe it is interesting to give a critical account of our entire work in this area and justify the statement “to label means to change”. New perspectives regarding combined therapeutic approaches and the use of rhodamine B conjugates to target closely related problems such as bacterial resistance and biofilm production are also discussed.
Collapse
|
14
|
Leitch HA, Buckstein R, Zhu N, Nevill TJ, Yee KWL, Leber B, Keating MM, St Hilaire E, Kumar R, Delage R, Geddes M, Storring JM, Shamy A, Elemary M, Wells RA. Iron overload in myelodysplastic syndromes: Evidence based guidelines from the Canadian consortium on MDS. Leuk Res 2018; 74:21-41. [PMID: 30286330 DOI: 10.1016/j.leukres.2018.09.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 08/08/2018] [Accepted: 09/15/2018] [Indexed: 01/19/2023]
Abstract
In 2008 the first evidence-based Canadian consensus guideline addressing the diagnosis, monitoring and management of transfusional iron overload in patients with myelodysplastic syndromes (MDS) was published. The Canadian Consortium on MDS, comprised of hematologists from across Canada with a clinical and academic interest in MDS, reconvened to update these guidelines. A literature search was updated in 2017; topics reviewed include mechanisms of iron overload induced cellular damage, evidence for clinical endpoints impacted by iron overload including organ dysfunction, infections, marrow failure, overall survival, acute myeloid leukemia progression, and endpoints around hematopoietic stem-cell transplant. Evidence for an impact of iron reduction on the same endpoints is discussed, guidelines are updated, and areas identified where evidence is suboptimal. The guidelines address common questions around the diagnosis, workup and management of iron overload in clinical practice, and take the approach of who, when, why and how to treat iron overload in MDS. Practical recommendations for treatment and monitoring are made. Evidence levels and grading of recommendations are provided for all clinical endpoints examined.
Collapse
Affiliation(s)
- Heather A Leitch
- Hematology, St. Paul's Hospital and the University of British Columbia, Vancouver, BC, Canada.
| | - Rena Buckstein
- Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Nancy Zhu
- Hematology/Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Thomas J Nevill
- Leukemia/BMT Program of British Columbia, Division of Hematology, Vancouver, BC, Canada
| | - Karen W L Yee
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Brian Leber
- McMaster University, Hamilton, Ontario, Canada
| | | | - Eve St Hilaire
- Centre d'Oncologie, Dr-Leon-Richard, Moncton, New Brunswick, Canada
| | - Rajat Kumar
- Hematology/Oncology, CancerCare Manitoba, Winnipeg, Manitoba, Canada
| | - Robert Delage
- Hematology Department, Centre Hospitalier Universitaire, Laval University, Quebec, QC, Canada
| | - Michelle Geddes
- Department of Medicine/Hematology, Foothills Medical Centre, Calgary, Alberta, Canada
| | | | - April Shamy
- Sir Mortimer B Davis Hospital, McGill University, Montreal, Quebec, Canada
| | - Mohamed Elemary
- Saskatoon Cancer Center, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Richard A Wells
- Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| |
Collapse
|
15
|
Gomes AC, Moreira AC, Mesquita G, Gomes MS. Modulation of Iron Metabolism in Response to Infection: Twists for All Tastes. Pharmaceuticals (Basel) 2018; 11:ph11030084. [PMID: 30200471 PMCID: PMC6161156 DOI: 10.3390/ph11030084] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 08/27/2018] [Accepted: 08/28/2018] [Indexed: 12/21/2022] Open
Abstract
Iron is an essential nutrient for almost all living organisms, but is not easily made available. Hosts and pathogens engage in a fight for the metal during an infection, leading to major alterations in the host’s iron metabolism. Important pathological consequences can emerge from the mentioned interaction, including anemia. Several recent reports have highlighted the alterations in iron metabolism caused by different types of infection, and several possible therapeutic strategies emerge, based on the targeting of the host’s iron metabolism. Here, we review the most recent literature on iron metabolism alterations that are induced by infection, the consequent development of anemia, and the potential therapeutic approaches to modulate iron metabolism in order to correct iron-related pathologies and control the ongoing infection.
Collapse
Affiliation(s)
- Ana Cordeiro Gomes
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
- Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.
| | - Ana C Moreira
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
- Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.
| | - Gonçalo Mesquita
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
- Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.
| | - Maria Salomé Gomes
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
- Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.
- Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
16
|
Wong CAC, Wong SAY, Leitch HA. Iron overload in lower international prognostic scoring system risk patients with myelodysplastic syndrome receiving red blood cell transfusions: Relation to infections and possible benefit of iron chelation therapy. Leuk Res 2018; 67:75-81. [PMID: 29477023 DOI: 10.1016/j.leukres.2018.02.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 02/03/2018] [Accepted: 02/06/2018] [Indexed: 01/27/2023]
Abstract
BACKGROUND An increased incidence of infections and infectious mortality has been reported in myelodysplastic syndromes (MDS) patients receiving red blood cell (RBC) transfusions. METHODS We examined incidence of infections requiring antibiotics, antifungal or antiviral medications in transfused lower International Prognostic Scoring System (IPSS) risk MDS patients and whether this differed with iron chelation therapy (ICT). RESULTS 138 transfused MDS patients were lower IPSS risk. 59 received ICT; median duration was 13 months. There was no significant difference between groups in neutrophil count at first RBC transfusion or first infection. Infections included: bacterial, n = 88; viral; fungal; and mycobacterial; n = 2 each. In ICT and non-ICT patients, respectively, infections were (number [%]): patients, 23 (40.0%) and 22 (27.8%); episodes (median [range]), 2 (1-6) and 2 (1-5); hospitalizations, 16 (27.1%) and 8 (10.1%); and deaths, 0 (0%) and 1 (1.3%), p = NS for all. Median overall survival (OS) from first RBC transfusion was superior in ICT patients, p = 0.01, and remained significant in a multivariate analysis (MVA), p = 0.003. Median time to first infection (TTI) was 27 and 7.8 months, respectively, p < 0.0001, and ICT remained significant for TTI in an MVA, p = 0.02, hazard ratio 0.3. For ICT patients with blast count <5%, TTI was significantly superior (p = 0.004). CONCLUSIONS In this retrospective analysis, for lower IPSS risk MDS patients receiving RBC transfusions, though number and type of infections were similar between groups and despite similar neutrophil counts, time to first infection was significantly longer in ICT patients (p < 0.0001). These results should be confirmed in larger, prospective analyses.
Collapse
Affiliation(s)
| | | | - Heather A Leitch
- Division of Hematology, St. Paul's Hospital, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
17
|
Wu Q, Shen Y, Tao Y, Wei J, Wang H, An P, Zhang Z, Gao H, Zhou T, Wang F, Min J. Hemojuvelin regulates the innate immune response to peritoneal bacterial infection in mice. Cell Discov 2017; 3:17028. [PMID: 28815056 PMCID: PMC5556331 DOI: 10.1038/celldisc.2017.28] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 07/13/2017] [Indexed: 12/20/2022] Open
Abstract
Hereditary hemochromatosis and iron imbalance are associated with susceptibility to bacterial infection; however, the underlying mechanisms are poorly understood. Here, we performed in vivo bacterial infection screening using several mouse models of hemochromatosis, including Hfe (Hfe−/−), hemojuvelin (Hjv−/−), and macrophage-specific ferroportin-1 (Fpn1fl/fl;LysM-Cre+) knockout mice. We found that Hjv−/− mice, but not Hfe−/− or Fpn1fl/fl;LysM-Cre+ mice, are highly susceptible to peritoneal infection by both Gram-negative and Gram-positive bacteria. Interestingly, phagocytic cells in the peritoneum of Hjv−/− mice have reduced bacterial clearance, IFN-γ secretion, and nitric oxide production; in contrast, both cell migration and phagocytosis are normal. Expressing Hjv in RAW264.7 cells increased the level of phosphorylated Stat1 and nitric oxide production. Moreover, macrophage-specific Hjv knockout mice are susceptible to bacterial infection. Finally, we found that Hjv facilitates the secretion of IFN-γ via the IL-12/Jak2/Stat4 signaling pathway. Together, these findings reveal a novel protective role of Hjv in the early stages of antimicrobial defense.
Collapse
Affiliation(s)
- Qian Wu
- The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Zhejiang University, Hangzhou, China.,Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Yuanyuan Shen
- The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yunlong Tao
- The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jiayu Wei
- The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hao Wang
- The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Zhejiang University, Hangzhou, China.,Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Peng An
- The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhuzhen Zhang
- The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hong Gao
- The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Zhejiang University, Hangzhou, China
| | - Tianhua Zhou
- The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Zhejiang University, Hangzhou, China
| | - Fudi Wang
- The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Zhejiang University, Hangzhou, China.,Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
18
|
Masumi A, Mochida K, Takizawa K, Mizukami T, Kuramitsu M, Tsuruhara M, Mori S, Shibayama K, Yamaguchi K, Hamaguchi I. Mycobacterium avium infection induces the resistance of the interferon-γ response in mouse spleen cells at late stages of infection. Inflamm Regen 2016; 36:21. [PMID: 29259694 PMCID: PMC5725973 DOI: 10.1186/s41232-016-0024-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 08/02/2016] [Indexed: 11/14/2022] Open
Abstract
Background Bacterial infections cause an increase in the population of hematopoietic stem cells (HSCs). To investigate the downstream factors associated with hematopoietic stem cells, mice are infected with Mycobacterium avium (M. avium). Results Mycobacterium avium (M. avium) infection induces the enlargement of the spleen and changes in histopathology, including changes to the lineage populations. A dramatic expansion of Lin−c-kit+Sca-1+ (KSL) cells in mouse bone marrow cells and spleen cells was detected 4 weeks after infection with M. avium; however, there was no difference in the engraft activity between infected and un-infected mouse bone marrow cells. We tested the cytokine and cytokine-related gene expression after M. avium infection and found that IFN-γ expression increased and peaked at 4 weeks in both bone marrow and spleen cells. The expression of Sca-1 gene peaked at 4 weeks in the bone marrow but peaked at 2 weeks in spleen cells, although the Sca-1 surface marker peaked at 4 weeks after infection in both bone marrow and spleen cells. Interferon regulatory factor-2 (IRF-2) expression did not change in the bone marrow cells, whereas it decreased in spleen cells at 4 weeks and IRF-1 expression was up-regulated in both bone marrow and spleen cells after infection. However, the up-regulation of IRF-1 was not correlated with IFN-γ expression in the M. avium-infected mouse spleen cells. Conclusions This finding suggests that the IFN-γ production mediated by M. avium infection alters the population of KSL cells during host defense, and the down-regulation of the IFN-γ response in spleen cells occurs at the late stage after M. avium infection.
Collapse
Affiliation(s)
- Atsuko Masumi
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, Tokyo, Japan.,Present address: Faculty of Pharmaceutical Sciences, Aomori University, 2-3-1, Kohbata, Aomori-shi, Aomori, 030-0943 Japan
| | - Keiko Mochida
- Department of Bacteriology II, National Institute of Infectious Diseases, 4-7-1, Gakuen Musashimurayama-shi, Tokyo, 208-0011 Japan
| | - Kazuya Takizawa
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, Tokyo, Japan
| | - Takuo Mizukami
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, Tokyo, Japan
| | - Madoka Kuramitsu
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, Tokyo, Japan
| | - Momoka Tsuruhara
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, Tokyo, Japan
| | - Shigetarou Mori
- Department of Bacteriology II, National Institute of Infectious Diseases, 4-7-1, Gakuen Musashimurayama-shi, Tokyo, 208-0011 Japan
| | - Keigo Shibayama
- Department of Bacteriology II, National Institute of Infectious Diseases, 4-7-1, Gakuen Musashimurayama-shi, Tokyo, 208-0011 Japan
| | - Kazunari Yamaguchi
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, Tokyo, Japan
| | - Isao Hamaguchi
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, Tokyo, Japan
| |
Collapse
|
19
|
Abstract
Hepcidin is the master regulator of iron homeostasis in vertebrates. The synthesis of hepcidin is induced by systemic iron levels and by inflammatory stimuli. While the role of hepcidin in iron regulation is well established, its contribution to host defense is emerging as complex and multifaceted. In this review, we summarize the literature on the role of hepcidin as a mediator of antimicrobial immunity. Hepcidin induction during infection causes depletion of extracellular iron, which is thought to be a general defense mechanism against many infections by withholding iron from invading pathogens. Conversely, by promoting iron sequestration in macrophages, hepcidin may be detrimental to cellular defense against certain intracellular infections, although critical in vivo studies are needed to confirm this concept. It is not yet clear whether hepcidin exerts any iron-independent effects on host defenses.
Collapse
Affiliation(s)
- Kathryn Michels
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Elizabeta Nemeth
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California, United States of America
| | - Tomas Ganz
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California, United States of America
- Department of Pathology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California, United States of America
| | - Borna Mehrad
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, United States of America
- The Carter Center for Immunology, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Internal Medicine, University of Virginia, Charlottesville, Virginia, United States of America
- * E-mail:
| |
Collapse
|
20
|
Mycobacterium avium Complex Infection in a Patient with Sickle Cell Disease and Severe Iron Overload. Case Rep Infect Dis 2014; 2014:405323. [PMID: 25544913 PMCID: PMC4269307 DOI: 10.1155/2014/405323] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 10/28/2014] [Accepted: 11/06/2014] [Indexed: 12/03/2022] Open
Abstract
A 34-year-old female with sickle cell anemia (hemoglobin SS disease) and severe iron overload presented to our institution with the subacute presentation of recurrent pain crisis, fever of unknown origin, pancytopenia, and weight loss. A CT scan demonstrated both lung and liver nodules concerning for granulomatous disease. Subsequent biopsies of the liver and bone marrow confirmed the presence of noncaseating granulomas and blood cultures isolated Mycobacterium avium complex MAC. Disseminated MAC is considered an opportunistic infection typically diagnosed in the immunocompromised and rarely in immunocompetent patients. An appreciable number of mycobacterial infection cases have been reported in sickle cell disease patients without immune dysfunction. It has been reported that iron overload is known to increase the risk for mycobacterial infection in vitro and in vivo studies. While iron overload is primarily known to cause end organ dysfunction, the clinical relationship with sickle cell disease and disseminated MAC infection has not been reported. Clinical iron overload is a common condition diagnosed in the sub-Saharan African population. High dietary iron, genetic defects in iron trafficking, as well as hemoglobinopathy are believed to be the etiologies for iron overload in this region. Patients with iron overload in this region were 17-fold more likely to die from Mycobacterium tuberculosis. Both experimental and clinical evidence suggest a possible link to iron overload and mycobacterial infections; however larger observational studies are necessary to determine true causality.
Collapse
|
21
|
The ESAT-6 protein of Mycobacterium tuberculosis interacts with beta-2-microglobulin (β2M) affecting antigen presentation function of macrophage. PLoS Pathog 2014. [PMID: 25356553 DOI: 10.1371/journal.ppat.1004446.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
ESAT-6, an abundantly secreted protein of Mycobacterium tuberculosis (M. tuberculosis) is an important virulence factor, inactivation of which leads to reduced virulence of M. tuberculosis. ESAT-6 alone, or in complex with its chaperone CFP-10 (ESAT-6:CFP-10), is known to modulate host immune responses; however, the detailed mechanisms are not well understood. The structure of ESAT-6 or ESAT-6:CFP-10 complex does not suggest presence of enzymatic or DNA-binding activities. Therefore, we hypothesized that the crucial role played by ESAT-6 in the virulence of mycobacteria could be due to its interaction with some host cellular factors. Using a yeast two-hybrid screening, we identified that ESAT-6 interacts with the host protein beta-2-microglobulin (β2M), which was further confirmed by other assays, like GST pull down, co-immunoprecipitation and surface plasmon resonance. The C-terminal six amino acid residues (90-95) of ESAT-6 were found to be essential for this interaction. ESAT-6, in complex with CFP-10, also interacts with β2M. We found that ESAT-6/ESAT-6:CFP-10 can enter into the endoplasmic reticulum where it sequesters β2M to inhibit cell surface expression of MHC-I-β2M complexes, resulting in downregulation of class I-mediated antigen presentation. Interestingly, the ESAT-6:β2M complex could be detected in pleural biopsies of individuals suffering from pleural tuberculosis. Our data highlight a novel mechanism by which M. tuberculosis may undermine the host adaptive immune responses to establish a successful infection. Identification of such novel interactions may help us in designing small molecule inhibitors as well as effective vaccine design against tuberculosis.
Collapse
|
22
|
Sreejit G, Ahmed A, Parveen N, Jha V, Valluri VL, Ghosh S, Mukhopadhyay S. The ESAT-6 protein of Mycobacterium tuberculosis interacts with beta-2-microglobulin (β2M) affecting antigen presentation function of macrophage. PLoS Pathog 2014; 10:e1004446. [PMID: 25356553 PMCID: PMC4214792 DOI: 10.1371/journal.ppat.1004446] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 09/04/2014] [Indexed: 11/28/2022] Open
Abstract
ESAT-6, an abundantly secreted protein of Mycobacterium tuberculosis (M. tuberculosis) is an important virulence factor, inactivation of which leads to reduced virulence of M. tuberculosis. ESAT-6 alone, or in complex with its chaperone CFP-10 (ESAT-6:CFP-10), is known to modulate host immune responses; however, the detailed mechanisms are not well understood. The structure of ESAT-6 or ESAT-6:CFP-10 complex does not suggest presence of enzymatic or DNA-binding activities. Therefore, we hypothesized that the crucial role played by ESAT-6 in the virulence of mycobacteria could be due to its interaction with some host cellular factors. Using a yeast two-hybrid screening, we identified that ESAT-6 interacts with the host protein beta-2-microglobulin (β2M), which was further confirmed by other assays, like GST pull down, co-immunoprecipitation and surface plasmon resonance. The C-terminal six amino acid residues (90–95) of ESAT-6 were found to be essential for this interaction. ESAT-6, in complex with CFP-10, also interacts with β2M. We found that ESAT-6/ESAT-6:CFP-10 can enter into the endoplasmic reticulum where it sequesters β2M to inhibit cell surface expression of MHC-I-β2M complexes, resulting in downregulation of class I-mediated antigen presentation. Interestingly, the ESAT-6:β2M complex could be detected in pleural biopsies of individuals suffering from pleural tuberculosis. Our data highlight a novel mechanism by which M. tuberculosis may undermine the host adaptive immune responses to establish a successful infection. Identification of such novel interactions may help us in designing small molecule inhibitors as well as effective vaccine design against tuberculosis. M. tuberculosis is a dangerous and highly successful pathogen that has evolved several mechanisms to manipulate the host immune regulatory network. Proteins secreted by M. tuberculosis play important roles in virulence. One such protein is ESAT-6, which is secreted along with its chaperone CFP-10. Despite a host of studies highlighting modulation of immune responses by ESAT-6, there have not been many that identified host proteins interacting with ESAT-6. We have now found that the host protein β2M interacts very specifically with ESAT-6 at its C-terminal region. The soluble ESAT-6:CFP-10 complex was found to be trafficked into the endoplasmic reticulum, and treatment with recombinant ESAT-6:CFP-10 or the over-expression of ESAT-6 reduced cell surface expression of β2M and molecules which remain associated with it like HLA-I. Recombinant ESAT-6:CFP-10 was also found to reduce classical and cross presentation of peptide antigens by MHC-I molecules. In summary, our data indicate that interaction between ESAT-6 and β2M can reduce the levels of available free β2M that associate with HLA/MHC-I molecules. This could be an interesting mechanism by which M. tuberculosis inhibits classical and cross presentation of peptide antigens in order to prevent or delay the onset of anti-mycobacterial adaptive immune responses.
Collapse
Affiliation(s)
- Gopalkrishna Sreejit
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD), Nampally, Hyderabad, India
| | - Asma Ahmed
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD), Nampally, Hyderabad, India
| | - Nazia Parveen
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD), Nampally, Hyderabad, India
| | - Vishwanath Jha
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD), Nampally, Hyderabad, India
| | - Vijaya Lakshmi Valluri
- Division of Immunology and Molecular Biology, LEPRA Society-Blue Peter Research Centre, Hyderabad, India
| | - Sudip Ghosh
- Molecular Biology Unit, National Institute of Nutrition (ICMR), Jamai-Osmania Hyderabad, India
| | - Sangita Mukhopadhyay
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD), Nampally, Hyderabad, India
- * E-mail: ,
| |
Collapse
|
23
|
Nairz M, Schroll A, Demetz E, Tancevski I, Theurl I, Weiss G. 'Ride on the ferrous wheel'--the cycle of iron in macrophages in health and disease. Immunobiology 2014; 220:280-94. [PMID: 25240631 DOI: 10.1016/j.imbio.2014.09.010] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 08/20/2014] [Accepted: 09/05/2014] [Indexed: 12/16/2022]
Abstract
Iron homeostasis and macrophage biology are closely interconnected. On the one hand, iron exerts multiple effects on macrophage polarization and functionality. On the other hand, macrophages are central for mammalian iron homeostasis. The phagocytosis of senescent erythrocytes and their degradation by macrophages enable efficient recycling of iron and the maintenance of systemic iron balance. Macrophages express multiple molecules and proteins for the acquisition and utilization of iron and many of these pathways are affected by inflammatory signals. Of note, iron availability within macrophages has significant effects on immune effector functions and metabolic pathways within these cells. This review summarizes the physiological and pathophysiological aspects of macrophage iron metabolism and highlights its relevant consequences on immune function and in common diseases such as infection and atherosclerosis.
Collapse
Affiliation(s)
- Manfred Nairz
- Department of Internal Medicine VI, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Austria.
| | - Andrea Schroll
- Department of Internal Medicine VI, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Austria
| | - Egon Demetz
- Department of Internal Medicine VI, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Austria
| | - Ivan Tancevski
- Department of Internal Medicine VI, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Austria
| | - Igor Theurl
- Department of Internal Medicine VI, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Austria
| | - Günter Weiss
- Department of Internal Medicine VI, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Austria.
| |
Collapse
|
24
|
Oliveira F, Rocha S, Fernandes R. Iron metabolism: from health to disease. J Clin Lab Anal 2014; 28:210-8. [PMID: 24478115 DOI: 10.1002/jcla.21668] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2012] [Accepted: 07/24/2013] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Iron is vital for almost all living organisms by participating in a wide range of metabolic processes. However, iron concentration in body tissues must be tightly regulated since excessive iron may lead to microbial infections or cause tissue damage. Disorders of iron metabolism are among the most common human diseases and cover several conditions with varied clinical manifestations. METHODS An extensive literature review on the basic aspects of iron metabolism was performed, and the most recent findings on this field were highlighted as well. RESULTS New insights on iron metabolism have shed light into its real complexity, and its role in both healthy and pathological states has been recognized. Important discoveries about the iron regulatory machine and imbalances in its regulation have been made, which may lead in a near future to the development of new therapeutic strategies against iron disorders. Besides, the toxicity of free iron and its association with several pathologies has been addressed, although it requires further investigations. CONCLUSION This review will provide students in the fields of biochemistry and health sciences a brief and clear overview of iron physiology and toxicity, as well as imbalances in the iron homeostasis and associated pathological conditions.
Collapse
Affiliation(s)
- Fernando Oliveira
- Ciências Químicas e das Biomoléculas e Unidade de Mecanismos Moleculares da Doença do Centro de Investigação em Saúde e Ambiente, Escola Superior de Tecnologia da Saúde do Porto, Instituto Politécnico do Porto, Portugal
| | | | | |
Collapse
|
25
|
Silva-Gomes S, Vale-Costa S, Appelberg R, Gomes MS. Iron in intracellular infection: to provide or to deprive? Front Cell Infect Microbiol 2013; 3:96. [PMID: 24367768 PMCID: PMC3856365 DOI: 10.3389/fcimb.2013.00096] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 11/21/2013] [Indexed: 12/16/2022] Open
Abstract
Due to their chemical versatility, transition metals were incorporated as cofactors for several basic metabolic pathways in living organisms. This same characteristic makes them potentially harmful, since they can be engaged in deleterious reactions like Fenton chemistry. As such, organisms have evolved highly specialized mechanisms to supply their own metal needs while keeping their toxic potential in check. This dual character comes into play in host-pathogen interactions, given that the host can either deprive the pathogen of these key nutrients or exploit them to induce toxicity toward the invading agent. Iron stands as the prototypic example of how a metal can be used to limit the growth of pathogens by nutrient deprivation, a mechanism widely studied in Mycobacterium infections. However, the host can also take advantage of iron-induced toxicity to control pathogen proliferation, as observed in infections caused by Leishmania. Whether we may harness either of the two pathways for therapeutical purposes is still ill-defined. In this review, we discuss how modulation of the host iron availability impacts the course of infections, focusing on those caused by two relevant intracellular pathogens, Mycobacterium and Leishmania.
Collapse
Affiliation(s)
- Sandro Silva-Gomes
- Infection and Immunity Unit, Instituto de Biologia Molecular e Celular, Universidade do Porto Porto, Portugal ; Department of Molecular Biology, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto Porto, Portugal
| | - Sílvia Vale-Costa
- Infection and Immunity Unit, Instituto de Biologia Molecular e Celular, Universidade do Porto Porto, Portugal ; Department of Molecular Biology, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto Porto, Portugal
| | - Rui Appelberg
- Infection and Immunity Unit, Instituto de Biologia Molecular e Celular, Universidade do Porto Porto, Portugal ; Department of Molecular Biology, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto Porto, Portugal
| | - Maria S Gomes
- Infection and Immunity Unit, Instituto de Biologia Molecular e Celular, Universidade do Porto Porto, Portugal ; Department of Molecular Biology, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto Porto, Portugal
| |
Collapse
|
26
|
Silva-Gomes S, Bouton C, Silva T, Santambrogio P, Rodrigues P, Appelberg R, Gomes MS. Mycobacterium avium infection induces H-ferritin expression in mouse primary macrophages by activating Toll-like receptor 2. PLoS One 2013; 8:e82874. [PMID: 24349383 PMCID: PMC3857292 DOI: 10.1371/journal.pone.0082874] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 11/06/2013] [Indexed: 11/18/2022] Open
Abstract
Important for both host and pathogen survivals, iron is a key factor in determining the outcome of an infectious process. Iron with-holding, including sequestration inside tissue macrophages, is considered an important strategy to fight infection. However, for intra-macrophagic pathogens, such as Mycobacterium avium, host defence may depend on intracellular iron sequestration mechanisms. Ferritin, the major intracellular iron storage protein, plays a critical role in this process. In the current study, we studied ferritin expression in mouse bone marrow-derived macrophages upon infection with M. avium. We found that H-ferritin is selectively increased in infected macrophages, through an up-regulation of gene transcription. This increase was mediated by the engagement of Toll like receptor-2, and was independent of TNF-alpha or nitric oxide production. The formation of H-rich ferritin proteins and the consequent iron sequestration may be an important part of the panoply of antimicrobial mechanisms of macrophages.
Collapse
Affiliation(s)
- Sandro Silva-Gomes
- IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- ICBAS, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Cécile Bouton
- Institut de Chimie des Substances Naturelles, UPR2301 CNRS, Centre de Recherche de Gif, Gif-sur-Yvette, France
| | - Tânia Silva
- IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- ICBAS, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | | | - Pedro Rodrigues
- IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- ICBAS, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Rui Appelberg
- IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- ICBAS, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Maria Salomé Gomes
- IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- ICBAS, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
- * E-mail:
| |
Collapse
|
27
|
Heme catabolism by heme oxygenase-1 confers host resistance to Mycobacterium infection. Infect Immun 2013; 81:2536-45. [PMID: 23630967 DOI: 10.1128/iai.00251-13] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Heme oxygenases (HO) catalyze the rate-limiting step of heme degradation. The cytoprotective action of the inducible HO-1 isoform, encoded by the Hmox1 gene, is required for host protection against systemic infections. Here we report that upregulation of HO-1 expression in macrophages (M) is strictly required for protection against mycobacterial infection in mice. HO-1-deficient (Hmox1(-/-)) mice are more susceptible to intravenous Mycobacterium avium infection, failing to mount a protective granulomatous response and developing higher pathogen loads, than infected wild-type (Hmox1(+/+)) controls. Furthermore, Hmox1(-/-) mice also develop higher pathogen loads and ultimately succumb when challenged with a low-dose aerosol infection with Mycobacterium tuberculosis. The protective effect of HO-1 acts independently of adaptive immunity, as revealed in M. avium-infected Hmox1(-/-) versus Hmox1(+/+) SCID mice lacking mature B and T cells. In the absence of HO-1, heme accumulation acts as a cytotoxic pro-oxidant in infected M, an effect mimicked by exogenous heme administration to M. avium-infected wild-type M in vitro or to mice in vivo. In conclusion, HO-1 prevents the cytotoxic effect of heme in M, contributing critically to host resistance to Mycobacterium infection.
Collapse
|
28
|
Vale-Costa S, Gomes-Pereira S, Teixeira CM, Rosa G, Rodrigues PN, Tomás A, Appelberg R, Gomes MS. Iron overload favors the elimination of Leishmania infantum from mouse tissues through interaction with reactive oxygen and nitrogen species. PLoS Negl Trop Dis 2013; 7:e2061. [PMID: 23459556 PMCID: PMC3573095 DOI: 10.1371/journal.pntd.0002061] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 01/02/2013] [Indexed: 02/07/2023] Open
Abstract
Iron plays a central role in host-parasite interactions, since both intervenients need iron for survival and growth, but are sensitive to iron-mediated toxicity. The host's iron overload is often associated with susceptibility to infection. However, it has been previously reported that iron overload prevented the growth of Leishmania major, an agent of cutaneous leishmaniasis, in BALB/c mice. In order to further clarify the impact of iron modulation on the growth of Leishmania in vivo, we studied the effects of iron supplementation or deprivation on the growth of L. infantum, the causative agent of Mediterranean visceral leishmaniasis, in the mouse model. We found that dietary iron deficiency did not affect the protozoan growth, whereas iron overload decreased its replication in the liver and spleen of a susceptible mouse strain. The fact that the iron-induced inhibitory effect could not be seen in mice deficient in NADPH dependent oxidase or nitric oxide synthase 2 suggests that iron eliminates L. infantum in vivo through the interaction with reactive oxygen and nitrogen species. Iron overload did not significantly alter the mouse adaptive immune response against L. infantum. Furthermore, the inhibitory action of iron towards L. infantum was also observed, in a dose dependent manner, in axenic cultures of promastigotes and amastigotes. Importantly, high iron concentrations were needed to achieve such effects. In conclusion, externally added iron synergizes with the host's oxidative mechanisms of defense in eliminating L. infantum from mouse tissues. Additionally, the direct toxicity of iron against Leishmania suggests a potential use of this metal as a therapeutic tool or the further exploration of iron anti-parasitic mechanisms for the design of new drugs. Leishmania are important vector-borne protozoan pathogens that cause different forms of disease, ranging from cutaneous self-healing lesions to life-threatening visceral infection. L. infantum is the most common species causing visceral leishmaniasis in Europe and the Mediterranean basin. Iron plays a critical role in host-pathogen interactions. Both the microorganism and its host need iron for growth. However, iron may promote the formation of toxic reactive oxygen species, which contribute to pathogen elimination, but also to host tissue pathology. We investigated the effect of manipulating host iron status on the outcome of L. infantum infection, using the mouse as an experimental model. We found that dietary iron deprivation had no effect on L. infantum growth, and iron-dextran injection decreased the multiplication of L. infantum in mouse organs. The fact that this anti-parasitic effect of iron was not observed in mice genetically deficient in superoxide and nitric oxide synthesis pathways indicates that iron is likely to act in synergy with reactive oxygen and nitrogen species produced by the host's macrophages. This work clearly shows that iron supplementation improves the host's capacity to eliminate L. infantum parasites and suggests that iron may be further explored as a therapeutic tool to fight this type of infection.
Collapse
Affiliation(s)
- Sílvia Vale-Costa
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Sandra Gomes-Pereira
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- CISA-ESTSP - Núcleo de Investigação em Farmácia, Centro de Investigação em Saúde e Ambiente, Escola Superior de Tecnologia da Saúde do Porto, Instituto Politécnico do Porto, Porto, Portugal
| | - Carlos Miguel Teixeira
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Gustavo Rosa
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Pedro Nuno Rodrigues
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Ana Tomás
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Rui Appelberg
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Maria Salomé Gomes
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
- * E-mail:
| |
Collapse
|
29
|
Systemic and cellular consequences of macrophage control of iron metabolism. Semin Immunol 2012; 24:393-8. [DOI: 10.1016/j.smim.2013.01.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 01/07/2013] [Indexed: 01/25/2023]
|
30
|
Iron metabolism and the innate immune response to infection. Microbes Infect 2011; 14:207-16. [PMID: 22033148 DOI: 10.1016/j.micinf.2011.10.001] [Citation(s) in RCA: 185] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Revised: 09/29/2011] [Accepted: 10/10/2011] [Indexed: 12/15/2022]
Abstract
Host antimicrobial mechanisms reduce iron availability to pathogens. Iron proteins influencing the innate immune response include hepcidin, lactoferrin, siderocalin, haptoglobin, hemopexin, Nramp1, ferroportin and the transferrin receptor. Numerous global health threats are influenced by iron status and provide examples of our growing understanding of the connections between infection and iron metabolism.
Collapse
|
31
|
Rodrigues PN, Gomes SS, Neves JV, Gomes-Pereira S, Correia-Neves M, Nunes-Alves C, Stolte J, Sanchez M, Appelberg R, Muckenthaler MU, Gomes MS. Mycobacteria-induced anaemia revisited: A molecular approach reveals the involvement of NRAMP1 and lipocalin-2, but not of hepcidin. Immunobiology 2011; 216:1127-34. [DOI: 10.1016/j.imbio.2011.04.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2010] [Revised: 04/11/2011] [Accepted: 04/13/2011] [Indexed: 01/08/2023]
|
32
|
Abstract
My laboratory has been interested for some time in the influence of iron, a nutrient that is essential for both microbial pathogens and their mammalian hosts, on the course of infectious disease. Our studies indicate that alterations in the expression of host molecules that sequester or transport iron can have direct effects on pathogen growth and can also have an impact on the ability to mount normal immune responses. We have elucidated the mechanistic basis for some of these observations, and have started to apply our findings in strategies to control abnormalities of inflammation and iron metabolism. I will review here what we have learned about the interactions between iron and immunity and discuss the implications of the information that we have acquired.
Collapse
Affiliation(s)
- Bobby J Cherayil
- Mucosal Immunology Laboratory, MassGeneral Hospital for Children, Charlestown, MA 02129, USA.
| |
Collapse
|
33
|
Nairz M, Schroll A, Sonnweber T, Weiss G. The struggle for iron - a metal at the host-pathogen interface. Cell Microbiol 2010; 12:1691-702. [DOI: 10.1111/j.1462-5822.2010.01529.x] [Citation(s) in RCA: 283] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
34
|
Wang L, Cherayil BJ. Ironing out the wrinkles in host defense: interactions between iron homeostasis and innate immunity. J Innate Immun 2009; 1:455-64. [PMID: 20375603 PMCID: PMC3969595 DOI: 10.1159/000210016] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2008] [Revised: 01/16/2009] [Accepted: 01/16/2009] [Indexed: 12/15/2022] Open
Abstract
Iron is an essential micronutrient for both microbial pathogens and their mammalian hosts. Changes in iron availability and distribution have significant effects on pathogen virulence and on the immune response to infection. Recent advances in our understanding of the molecular regulation of iron metabolism have shed new light on how alterations in iron homeostasis both contribute to and influence innate immunity. In this article, we review what is currently known about the role of iron in the response to infection.
Collapse
Affiliation(s)
- Lijian Wang
- Mucosal Immunology Laboratory, Massachusetts General Hospital, Charlestown, Mass., USA
- Department of Nutrition, Harvard School of Public Health, Boston, Mass., USA
| | - Bobby J. Cherayil
- Mucosal Immunology Laboratory, Massachusetts General Hospital, Charlestown, Mass., USA
- Department of Pediatrics, Harvard Medical School, Boston, Mass., USA
| |
Collapse
|
35
|
Flórido M, Borges M, Rodrigues P, Vale-Costa S, Salomé Gomes M, Appelberg R. Constitutive expression of Bcl-2 in the haematopoietic compartment alters the metabolism of iron and increases resistance to mycobacterial infection. Clin Exp Immunol 2009; 156:61-8. [PMID: 19210523 DOI: 10.1111/j.1365-2249.2008.03867.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Mice expressing a vav-bcl-2 transgene were tested for their resistance to an experimental infection with Mycobacterium avium. When compared with control littermates, transgenic mice exhibited an increase in the resistance to infection which was independent of B or T lymphocytes and did not require the production of gamma interferon. Macrophages from both control and transgenic mice showed equal permissiveness to M. avium growth in vitro. Finally, transgenic mice expressed diminished circulating iron levels which correlated with the increased resistance to infection.
Collapse
Affiliation(s)
- M Flórido
- Laboratory of Microbiology and Immunology of Infection, Instituto de Biologia Molecular e Celular (IBMC), Portugal
| | | | | | | | | | | |
Collapse
|