1
|
Kim DH, Kim JH, Lim KB, Lee JB, Park SY, Song CS, Lee SW, Lee DH, Choi IS. Antiviral activity of adenoviral vector expressing human interferon lambda-4 against influenza virus. J Med Virol 2024; 96:e29605. [PMID: 38634474 DOI: 10.1002/jmv.29605] [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: 01/22/2024] [Revised: 03/13/2024] [Accepted: 04/04/2024] [Indexed: 04/19/2024]
Abstract
Interferon lambda (IFNλ), classified as a type III IFN, is a representative cytokine that plays an important role in innate immunity along with type I IFN. IFNλ can elicit antiviral states by inducing peculiar sets of IFN-stimulated genes (ISGs). In this study, an adenoviral vector expression system with a tetracycline operator system was used to express human IFNλ4 in cells and mice. The formation of recombinant adenovirus (rAd-huIFNλ4) was confirmed using immunohistochemistry assays and transmission electron microscopy. Its purity was verified by quantifying host cell DNA and host cell proteins, as well as by confirming the absence of the replication-competent adenovirus. The transduction of rAd-huIFNλ4 induced ISGs and inhibited four subtypes of the influenza virus in both mouse-derived (LA-4) and human-derived cells (A549). The antiviral state was confirmed in BALB/c mice following intranasal inoculation with 109 PFU of rAd-huIFNλ4, which led to the inhibition of four subtypes of the influenza virus in mouse lungs, with reduced inflammatory lesions. These results imply that human IFNλ4 could induce antiviral status by modulating ISG expression in mice.
Collapse
Affiliation(s)
- Dong-Hwi Kim
- Department of Infectious Diseases, College of Veterinary Medicine, Konkuk University, Gwangjin-gu, Seoul, Republic of Korea
| | - Jae-Hyeong Kim
- Department of Infectious Diseases, College of Veterinary Medicine, Konkuk University, Gwangjin-gu, Seoul, Republic of Korea
| | - Kyu-Beom Lim
- Department of Infectious Diseases, College of Veterinary Medicine, Konkuk University, Gwangjin-gu, Seoul, Republic of Korea
| | - Joong-Bok Lee
- Department of Infectious Diseases, College of Veterinary Medicine, Konkuk University, Gwangjin-gu, Seoul, Republic of Korea
- Konkuk University Zoonotic Diseases Research Center, Konkuk University, Gwangjin-gu, Seoul, Republic of Korea
| | - Seung-Yong Park
- Department of Infectious Diseases, College of Veterinary Medicine, Konkuk University, Gwangjin-gu, Seoul, Republic of Korea
- Konkuk University Zoonotic Diseases Research Center, Konkuk University, Gwangjin-gu, Seoul, Republic of Korea
| | - Chang-Seon Song
- Department of Infectious Diseases, College of Veterinary Medicine, Konkuk University, Gwangjin-gu, Seoul, Republic of Korea
- Konkuk University Zoonotic Diseases Research Center, Konkuk University, Gwangjin-gu, Seoul, Republic of Korea
| | - Sang-Won Lee
- Department of Infectious Diseases, College of Veterinary Medicine, Konkuk University, Gwangjin-gu, Seoul, Republic of Korea
- Konkuk University Zoonotic Diseases Research Center, Konkuk University, Gwangjin-gu, Seoul, Republic of Korea
| | - Dong-Hun Lee
- Department of Infectious Diseases, College of Veterinary Medicine, Konkuk University, Gwangjin-gu, Seoul, Republic of Korea
- Konkuk University Zoonotic Diseases Research Center, Konkuk University, Gwangjin-gu, Seoul, Republic of Korea
| | - In-Soo Choi
- Department of Infectious Diseases, College of Veterinary Medicine, Konkuk University, Gwangjin-gu, Seoul, Republic of Korea
- Konkuk University Zoonotic Diseases Research Center, Konkuk University, Gwangjin-gu, Seoul, Republic of Korea
- KU Center for Animal Blood Medical Science, Konkuk University, Gwangjin-gu, Seoul, Republic of Korea
| |
Collapse
|
2
|
Kim HI, Kim DS, Jung Y, Sung NY, Kim M, Han IJ, Nho EY, Hong JH, Lee JK, Boo M, Kim HL, Baik S, Jung KO, Lee S, Kim CS, Park J. Immune-Enhancing Effect of Sargassum horneri on Cyclophosphamide-Induced Immunosuppression in BALB/c Mice and Primary Cultured Splenocytes. Molecules 2022; 27:8253. [PMID: 36500343 PMCID: PMC9738764 DOI: 10.3390/molecules27238253] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/09/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
Sargassum horneri (SH) is a seaweed that has several features that benefit health. In this study, we investigated the immune-enhancing effect of SH, focusing on the role of spleen-mediated immune functions. Chromatographic analysis of SH identified six types of monosaccharide contents, including mannose, rhamnose glucose, galactose xylose and fucose. SH increased cell proliferation of primary cultured naïve splenocytes treated with or without cyclophosphamide (CPA), an immunosuppression agent. SH also reversed the CPA-induced decrease in Th1 cytokines. In vivo investigation revealed that SH administration can increase the tissue weight of major immune organs, such as the spleen and thymus. A similar effect was observed in CPA-injected immunosuppressed BALB/c mice. SH treatment increased the weight of the spleen and thymus, blood immune cell count and Th1 cytokine expression. Additionally, the YAC-1-targeting activities of natural killer cells, which are important in innate immunity, were upregulated upon SH treatment. Overall, our study demonstrates the immune-enhancing effect of SH, suggesting its potential as a medicinal or therapeutic agent for pathologic conditions involving immunosuppression.
Collapse
Affiliation(s)
- Hyo In Kim
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Dong-Sub Kim
- Division of Natural Product Research, Korea Prime Pharmacy Co., Ltd., Suwon 16229, Republic of Korea
| | - Yunu Jung
- Division of Natural Product Research, Korea Prime Pharmacy Co., Ltd., Suwon 16229, Republic of Korea
| | - Nak-Yun Sung
- Division of Natural Product Research, Korea Prime Pharmacy Co., Ltd., Suwon 16229, Republic of Korea
| | - Minjee Kim
- Division of Natural Product Research, Korea Prime Pharmacy Co., Ltd., Suwon 16229, Republic of Korea
| | - In-Jun Han
- Division of Natural Product Research, Korea Prime Pharmacy Co., Ltd., Suwon 16229, Republic of Korea
| | - Eun Yeong Nho
- Division of Natural Product Research, Korea Prime Pharmacy Co., Ltd., Suwon 16229, Republic of Korea
| | - Joon Ho Hong
- Nano Bio Research Center, Jeonnam Bioindustry Foundation, Jangsung 57248, Republic of Korea
| | - Jin-Kyu Lee
- Department of Food Regulatory Science, Korea University, Sejong 30019, Republic of Korea
| | - Mina Boo
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hye-Lin Kim
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sangyul Baik
- School of Mechanical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Kyung Oh Jung
- Department of Anatomy, College of Medicine, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Sanghyun Lee
- Department of Plant Science and Technology, Chung-Ang University, Anseong 17546, Republic of Korea
| | - Chun Sung Kim
- Department of Oral Biochemistry, College of Dentistry, Chosun University, Gwangju 61452, Republic of Korea
| | - Jinbong Park
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
3
|
McGonagle DG, McInnes IB, Kirkham BW, Sherlock J, Moots R. The role of IL-17A in axial spondyloarthritis and psoriatic arthritis: recent advances and controversies. Ann Rheum Dis 2019; 78:1167-1178. [PMID: 31278139 PMCID: PMC6788885 DOI: 10.1136/annrheumdis-2019-215356] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 05/17/2019] [Accepted: 06/14/2019] [Indexed: 12/19/2022]
Abstract
Although the pathogenic mechanisms underlying axial spondyloarthritis (axSpA) and psoriatic arthritis (PsA) are not fully elucidated, several lines of evidence suggest that immune responses mediated by interleukin 17A (IL-17A) play a pivotal role in both diseases. This is best highlighted by the significant clinical efficacy shown with inhibitors of IL-17A in treating axSpA and PsA. Nevertheless, a number of knowledge gaps exist regarding the role of IL-17A in the pathophysiology of spondyloarthritis in man, including its cellular origin, its precise role in discrete disease processes such enthesitis, bone erosion, and bone formation, and the reasons for the discrepant responses to IL-17A inhibition observed in certain other spondyloarthritis manifestations. In this review, we focus on the latest data from studies investigating the role of IL-17A in ankylosing spondylitis (AS) and PsA that build on existing and emerging scientific knowledge in the field. Key remaining research questions are also highlighted to guide future research.
Collapse
Affiliation(s)
- Dennis G McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.,NIHR Leeds Musculoskeletal Biomedical Research Unit, Chapel Allerton, Leeds Teaching Hospital Trust, Leeds, UK
| | - Iain B McInnes
- Institute of Infection Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Bruce W Kirkham
- Rheumatology Department, Guy's and Saint Thomas' NHS Foundation Trust, London, UK
| | - Jonathan Sherlock
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK.,Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Robert Moots
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK .,Department of Academic Rheumatology, Aintree University Hospital, Liverpool, UK
| |
Collapse
|
4
|
Oligbu G, Collins S, Andrews N, Sheppard CL, Fry NK, Slack MPE, Borrow R, Ladhani SN. Characteristics and Serotype Distribution of Childhood Cases of Invasive Pneumococcal Disease Following Pneumococcal Conjugate Vaccination in England and Wales, 2006-2014. Clin Infect Dis 2019; 65:1191-1198. [PMID: 29309553 DOI: 10.1093/cid/cix418] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 05/03/2017] [Indexed: 11/13/2022] Open
Abstract
Background The 7-valent and 13-valent pneumococcal conjugate vaccines (PCV7 and PCV13, respectively) are highly effective in preventing invasive pneumococcal disease (IPD) caused by vaccine serotypes. Vaccine failure (vaccine-type IPD after age-appropriate immunization) is rare. Little is known about the risk, clinical characteristics, or outcomes of PCV13 compared to PCV7 vaccine failure. Methods Public Health England conducts IPD surveillance and provides a national reference service for serotyping pneumococcal isolates in England and Wales. We compared the epidemiology, rates, risk factors, serotype distribution, clinical characteristics, and outcomes of IPD in children with PCV13 and PCV7 vaccine failure. Results A total of 163 episodes of PCV failure were confirmed in 161 children over 8 years (4 September 2006 to 3 September 2014) in 10 birth cohorts. After 3 vaccine doses, PCV7 and PCV13 failure rates were 0.19/100000 (95% confidence interval [CI], .10-.33 [57 cases]) and 0.66/100000 (95% CI, .44-.95 [104 cases]) vaccinated person-years, respectively. Children with PCV13 failure were more likely to be healthy (87/105 [82.9%] vs 37/56 [66.1%]; P = .02), present with bacteremic lower respiratory tract infection (LRTI) (61/105 [58.1%] vs 11/56 [19.6%]; P < .001), and develop empyema (41/61 [67.2%] vs 1/11 [9.1%]; P < .001) compared to PCV7 failures. Serotypes 3 (n = 38 [36.2%]) and 19A (n = 30 [28.6%]) were responsible for most PCV13 failures. Six children died (4% [95% CI, 1%-8%]), including 5 with comorbidities. Conclusions PCV failure is rare and, compared to PCV7 serotypes, the additional PCV13 serotypes are more likely to cause bacteremic LRTI and empyema in healthy vaccinated children.
Collapse
Affiliation(s)
- Godwin Oligbu
- Paediatric Infectious Diseases Research Group, Institute for Infection and Immunity, St George's, University of London
| | - Sarah Collins
- Immunisation, Hepatitis and Blood Safety Department, Public Health England, London, United Kingdom
| | - Nick Andrews
- Statistics, Modelling and Economics Department, Public Health England, London, United Kingdom
| | - Carmen L Sheppard
- Respiratory and Vaccine Preventable Bacterial Reference Unit, Public Health England, London, United Kingdom
| | - Norman K Fry
- Respiratory and Vaccine Preventable Bacterial Reference Unit, Public Health England, London, United Kingdom
| | - Mary P E Slack
- Institute of Hygiene and Microbiology, University of Wurzburg, Germany.,School of Medicine, Griffith University Gold Coast Campus, Southport, Queensland, Australia
| | - Ray Borrow
- Vaccine Evaluation Unit, Public Health England, Manchester Royal Infirmary, United Kingdom
| | - Shamez N Ladhani
- Paediatric Infectious Diseases Research Group, Institute for Infection and Immunity, St George's, University of London.,Immunisation, Hepatitis and Blood Safety Department, Public Health England, London, United Kingdom
| |
Collapse
|
5
|
Proctor RA. Immunity to Staphylococcus aureus: Implications for Vaccine Development. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0037-2018. [PMID: 31298209 PMCID: PMC10957185 DOI: 10.1128/microbiolspec.gpp3-0037-2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Indexed: 12/19/2022] Open
Abstract
Cell-mediated immunity seems to be critical for prevention and resolution of invasive S. aureus infections, but an imbalance in this immunity may also produce SIRS and death or an inadequate protective response with prolonged bacteremia and death. This dysregulation is likely at the heart of mortality and severe disease in humans. Anti-toxin antibodies may also come into play in reducing the severity of S. aureus infections, but these antibodies might also address superantigen-induced immune dysregulation. Thus, while changing intrinsic T cell responses may be therapeutically difficult, monoclonal antibodies against superantigens may have utility in addressing dysfunctional immune responses to S. aureus. The models above are hypotheses for examining, and potentially dramatically improving immune response to and safety of S. aureus vaccines.
Collapse
Affiliation(s)
- Richard A Proctor
- University of Wisconsin, Medical Microbiology/Immunology, Madison, WI 53705
| |
Collapse
|
6
|
Fayez EA, Koohini Z, Koohini Z, Zamanzadeh H, de Boer M, Roos D, Teimourian S. Characterization of two novel mutations in IL-12R signaling in MSMD patients. Pathog Dis 2019; 77:ftz030. [PMID: 31158284 DOI: 10.1093/femspd/ftz030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 05/31/2019] [Indexed: 12/17/2023] Open
Abstract
Mendelian Susceptibility to Mycobacterial Disease (MSMD) is a rare syndrome with infections-among other complications-after Bacillus Calmette-Guerin (BCG) vaccination in children. We focused on the IL-12/IFN-γ pathway to identify new mutations in our patients. This study included 20 patients by vulnerability to mycobacteria and clinical manifestations of severe, recurrent infections. Blood samples were activated with BCG, BCG + IL-12 and BCG + IFN-γ. Cytokine levels were analyzed by ELISA. Measurements of IL-12Rβ1 and IL-12Rβ2 on the surface of peripheral blood mononuclear cells were performed by flow cytometry. To detect genetic defects, next-generation sequencing was performed by Thermo Fisher immunodeficiency panel. Flow cytometry analysis of 20 patients indicated reduction in IL-12R (β1/β2) expression in seven patients who showed incomplete production of IFN-γ by ELISA. In the patient with reduced IL-12 production, IFN-γR and IL-12R (β1/β2) expression levels were normal. Mutation analysis showed three previously reported mutations, two novel mutations in IL-12 R (β1/β2), and one previously reported mutation in IL-12.
Collapse
Affiliation(s)
- Elham Alipour Fayez
- Department of Immunology, School of Medicine, Iran University of Medical Sciences Tehran, Iran
| | - Zahra Koohini
- Department of Medical Genetics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zohreh Koohini
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hossein Zamanzadeh
- Department of biology, School of basic sciences, University of Sistan and Balouchestan, Zahedan, Iran
| | - Martin de Boer
- Sanquin Research, and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Dirk Roos
- Sanquin Research, and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Shahram Teimourian
- Department of Medical Genetics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Schimke LF, Hibbard J, Martinez-Barricarte R, Khan TA, de Souza Cavalcante R, Borges de Oliveira Junior E, Takahashi França T, Iqbal A, Yamamoto G, Arslanian C, Feriotti C, Costa TA, Bustamante J, Boisson-Dupuis S, Casanova JL, Marzagao Barbuto JA, Zatz M, Poncio Mendes R, Garcia Calich VL, Ochs HD, Torgerson TR, Cabral-Marques O, Condino-Neto A. Paracoccidioidomycosis Associated With a Heterozygous STAT4 Mutation and Impaired IFN-γ Immunity. J Infect Dis 2019; 216:1623-1634. [PMID: 29029192 DOI: 10.1093/infdis/jix522] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 09/24/2017] [Indexed: 01/01/2023] Open
Abstract
Background Mutations in genes affecting interferon-γ (IFN-γ) immunity have contributed to understand the role of IFN-γ in protection against intracellular pathogens. However, inborn errors in STAT4, which controls interleukin-12 (IL-12) responses, have not yet been reported. Our objective was to determine the genetic defect in a family with a history of paracoccidioidomycosis. Methods Genetic analysis was performed by whole-exome sequencing and Sanger sequencing. STAT4 phosphorylation (pSTAT4) and translocation to the nucleus, IFN-γ release by patient lymphocytes, and microbicidal activity of patient monocytes/macrophages were assessed. The effect on STAT4 function was evaluated by site-directed mutagenesis using a lymphoblastoid B cell line (B-LCL) and U3A cells. Results A heterozygous missense mutation, c.1952 A>T (p.E651V) in STAT4 was identified in the index patient and her father. Patient's and father's lymphocytes showed reduced pSTAT4, nuclear translocation, and impaired IFN-γ production. Mutant B-LCL and U3A cells also displayed reduced pSTAT4. Patient's and father's peripheral blood mononuclear cells and macrophages demonstrated impaired fungicidal activity compared with those from healthy controls that improved in the presence of recombinant human IFN-γ, but not rhIL-12. Conclusion Our data suggest autosomal dominant STAT4 deficiency as a novel inborn error of IL-12-dependent IFN-γ immunity associated with susceptibility to paracoccidioidomycosis.
Collapse
Affiliation(s)
- Lena F Schimke
- Department of Immunology, University of Sao Paulo, Brazil.,Department of Rheumatology and Clinical Immunology, University of Lübeck, Germany
| | - James Hibbard
- Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Research Institute, New York
| | | | - Taj Ali Khan
- Department of Immunology, University of Sao Paulo, Brazil
| | | | | | | | - Asif Iqbal
- Laboratory of Biochemistry and Biophysics, Butantan Institute, Sao Paulo, Brazil
| | - Guilherme Yamamoto
- Human Genome and Stem Cell Research Center, University of Sao Paulo, Brazil
| | | | | | | | - Jacinta Bustamante
- St Giles Laboratory of Human Genetics of Infectious Diseases, the Rockefeller University, New York.,Imagine Institute, Paris Descartes University, Paris, France.,Laboratory of Human Genetics of Infectious Diseases, Paris, France.,Center for the Study of Primary Immunodeficiencies, Paris, France
| | - Stéphanie Boisson-Dupuis
- St Giles Laboratory of Human Genetics of Infectious Diseases, the Rockefeller University, New York.,Imagine Institute, Paris Descartes University, Paris, France.,Laboratory of Human Genetics of Infectious Diseases, Paris, France
| | - Jean-Laurent Casanova
- St Giles Laboratory of Human Genetics of Infectious Diseases, the Rockefeller University, New York.,Imagine Institute, Paris Descartes University, Paris, France.,Laboratory of Human Genetics of Infectious Diseases, Paris, France.,Pediatric Hematology-Immunology Unit, Necker Hospital for Sick Children, Paris, France.,Howard Hughes Medical Institute, the Rockefeller University, New York
| | | | - Mayana Zatz
- Human Genome and Stem Cell Research Center, University of Sao Paulo, Brazil
| | | | | | - Hans D Ochs
- Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Research Institute, New York
| | - Troy R Torgerson
- Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Research Institute, New York
| | - Otávio Cabral-Marques
- Department of Immunology, University of Sao Paulo, Brazil.,Department of Rheumatology and Clinical Immunology, University of Lübeck, Germany
| | - Antonio Condino-Neto
- Department of Immunology, University of Sao Paulo, Brazil.,Institute of Tropical Medicine, University of Sao Paulo, Brazil
| |
Collapse
|
8
|
Cul4 E3 ubiquitin ligase regulates ovarian cancer drug resistance by targeting the antiapoptotic protein BIRC3. Cell Death Dis 2019; 10:104. [PMID: 30718461 PMCID: PMC6362125 DOI: 10.1038/s41419-018-1200-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 10/30/2018] [Accepted: 11/02/2018] [Indexed: 02/05/2023]
Abstract
CRL4, a well-defined E3 ligase, has been reported to be upregulated and is proposed to be a potential drug target in ovarian cancers. However, the biological functions of CRL4 and the underlying mechanism regulating cancer chemoresistance are still largely elusive. Here, we show that CRL4 is considerably increased in cisplatin-resistant ovarian cancer cells, and CRL4 knockdown with shRNAs is able to reverse cisplatin-resistance of ovarian cancer cells. Moreover, CRL4 knockdown markedly inhibits the expression of BIRC3, one of the inhibitors of apoptosis proteins (IAPs). Besides, lower expression level of BIRC3 is associated with better prognosis of ovarian cancer patients, and BIRC3 knockdown in ovarian cancer cells can recover their sensitivity to cisplatin. More importantly, we demonstrate that CRL4 regulates BIRC3 expression by mediating the STAT3, but not the PI3K pathway. Therefore, our results identified CRL4 as an important factor in ovarian cancer chemoresistance, suggesting that CRL4 and BIRC3 may serve as novel therapeutic targets for relapsed patients after treatment with cisplatin and its derivative to overcome the bottle neck of ovarian cancer chemoresistance.
Collapse
|
9
|
Zhu F, Hu Y. Integrity of IKK/NF-κB Shields Thymic Stroma That Suppresses Susceptibility to Autoimmunity, Fungal Infection, and Carcinogenesis. Bioessays 2018. [PMID: 29522649 DOI: 10.1002/bies.201700131] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A pathogenic connection between autoreactive T cells, fungal infection, and carcinogenesis has been demonstrated in studies of human autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED) as well as in a mouse model in which kinase-dead Ikkα knock-in mice develop impaired central tolerance, autoreactive T cell-mediated autoimmunity, chronic fungal infection, and esophageal squamous cell carcinoma, which recapitulates APECED. IκB kinase α (IKKα) is one subunit of the IKK complex required for NF-κB activation. IKK/NF-κB is essential for central tolerance establishment by regulating the development of medullary thymic epithelial cells (mTECs) that facilitate the deletion of autoreactive T cells in the thymus. In this review, we extensively discuss the pathogenic roles of inborn errors in the IKK/NF-κB loci in the phenotypically related diseases APECED, immune deficiency syndrome, and severe combined immunodeficiency; differentiate how IKK/NF-κB components, through mTEC (stroma), T cells/leukocytes, or epithelial cells, contribute to the pathogenesis of infectious diseases, autoimmunity, and cancer; and highlight the medical significance of IKK/NF-κB in these diseases.
Collapse
Affiliation(s)
- Feng Zhu
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, 21701, Maryland, USA
| | - Yinling Hu
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, 21701, Maryland, USA
| |
Collapse
|
10
|
Gianchecchi E, Delfino DV, Fierabracci A. NK cells in autoimmune diseases: Linking innate and adaptive immune responses. Autoimmun Rev 2018; 17:142-154. [DOI: 10.1016/j.autrev.2017.11.018] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
11
|
Abstract
Interleukin (IL)-12 is composed of p35 and p40 subunits; in this case, IL-12p40 deficiency is a rare genetic etiology of Mendelian susceptibility to mycobacterial disease. Salmonellosis has been reported in almost half of these patients and mostly present in recurrent extraintestinal form. In this report, we described an 18-month-old boy with absence of IL-12p40 production suffering from chronic disseminated nontyphoidal salmonellosis. To the best of our knowledge, this is the first-reported case.
Collapse
|
12
|
Borghesi A, Stronati M, Fellay J. Neonatal Group B Streptococcal Disease in Otherwise Healthy Infants: Failure of Specific Neonatal Immune Responses. Front Immunol 2017; 8:215. [PMID: 28326082 PMCID: PMC5339282 DOI: 10.3389/fimmu.2017.00215] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 02/15/2017] [Indexed: 12/26/2022] Open
Abstract
Only a small proportion of newborn infants exposed to a pathogenic microorganism develop overt infection. Susceptibility to infection in preterm infants and infants with known comorbidities has a likely multifactorial origin and can be often attributed to the concurrence of iatrogenic factors, environmental determinants, underlying pathogenic processes, and probably genetic predisposition. Conversely, infection occurring in otherwise healthy full-term newborn infants is unexplained in most cases. Microbial virulence factors and the unique characteristics of the neonatal immune system only partially account for the interindividual variability in the neonatal immune responses to pathogens. We here suggest that neonatal infection occurring in otherwise healthy infants is caused by a failure of the specific protective immunity to the microorganism. To explain infection in term and preterm infants, we propose an extension of the previously proposed model of the genetic architecture of infectious diseases in humans. We then focus on group B streptococcus (GBS) disease, the best characterized neonatal infection, and outline the potential molecular mechanisms underlying the selective failure of the immune responses against GBS. In light of the recent discoveries of pathogen-specific primary immunodeficiencies and of the role of anticytokine autoantibodies in increasing susceptibility to specific infections, we hypothesize that GBS disease occurring in otherwise healthy infants could reflect an immunodeficiency caused either by rare genetic defects in the infant or by transmitted maternal neutralizing antibodies. These hypotheses are consistent with available epidemiological data, with clinical and epidemiological observations, and with the state of the art of neonatal physiology and disease. Studies should now be designed to comprehensively search for genetic or immunological factors involved in susceptibility to severe neonatal infections.
Collapse
Affiliation(s)
- Alessandro Borghesi
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Neonatal Intensive Care Unit, San Matteo Hospital, Pavia, Italy
| | - Mauro Stronati
- Neonatal Intensive Care Unit, San Matteo Hospital, Pavia, Italy
| | - Jacques Fellay
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| |
Collapse
|
13
|
Deng Y, Chang C, Lu Q. The Inflammatory Response in Psoriasis: a Comprehensive Review. Clin Rev Allergy Immunol 2017; 50:377-89. [PMID: 27025861 DOI: 10.1007/s12016-016-8535-x] [Citation(s) in RCA: 266] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Psoriasis is a chronic inflammatory autoimmune disease characterized by an excessively aberrant hyperproliferation of keratinocytes. The pathogenesis of psoriasis is complex and the exact mechanism remains elusive. However, psoriasis is thought to result from a combination of genetic, epigenetic, and environmental influences. Recent studies have identified that epigenetic factors including dysregulated DNA methylation levels, abnormal histone modification and microRNAs expressions are involved in the development of psoriasis. The interplay of immune cells and cytokines is another critical factor in the pathogenesis of psoriasis. These factors or pathways include Th1/Th2 homeostasis, the Th17/Treg balance and the IL-23/Th17 axis. Th17 is believed particularly important in psoriasis due to its pro-inflammatory effects and its involvement in an integrated inflammatory loop with dendritic cells and keratinocytes, contributing to an overproduction of antimicrobial peptides, inflammatory cytokines, and chemokines that leads to amplification of the immune response. In addition, other pathways and signaling molecules have been found to be involved, including Th9, Th22, regulatory T cells, γδ T cells, CD8(+) T cells, and their related cytokines. Understanding the pathogenesis of psoriasis will allow us to develop increasingly efficient targeted treatment by blocking relevant inflammatory signaling pathways and molecules. There is no cure for psoriasis at the present time, and much of the treatment involves managing the symptoms. The biologics, while lacking the adverse effects associated with some of the traditional medications such as corticosteroids and methotrexate, have their own set of side effects, which may include reactivation of latent infections. Significant challenges remain in developing safe and efficacious novel targeted therapies that depend on a better understanding of the immunological dysfunction in psoriasis.
Collapse
Affiliation(s)
- Yaxiong Deng
- Department of Dermatology, Second Xiangya Hospital, Hunan Key Laboratory of Medical Epigenomics, Central South University, Changsha, Hunan, China
| | - Christopher Chang
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis, 451 Health Sciences Drive, Suite 6510, Davis, CA, 95616, USA
| | - Qianjin Lu
- Department of Dermatology, Second Xiangya Hospital, Hunan Key Laboratory of Medical Epigenomics, Central South University, Changsha, Hunan, China. .,Second Xiangya Hospital, Central South University, #139 Renmin Middle Rd, Changsha, Hunan, 410011, China.
| |
Collapse
|
14
|
Mortaz E, Adcock IM, Tabarsi P, Darazam IA, Movassaghi M, Garssen J, Jamaati H, Velayati A. Pattern recognitions receptors in immunodeficiency disorders. Eur J Pharmacol 2017; 808:49-56. [PMID: 28095323 DOI: 10.1016/j.ejphar.2017.01.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Revised: 01/04/2017] [Accepted: 01/13/2017] [Indexed: 01/13/2023]
Abstract
Pattern recognition receptors (PRRs) recognize common microbial or host-derived macromolecules and have important roles in early activation and response of the immune system. Initiation of the innate immune response starts with the recognition of microbial structures called pathogen associated molecular patterns (PAMPs). Recognition of PAMPs is performed by germline-encoded receptors expressed mainly on immune cells termed pattern recognition receptors (PRRs). Several classes of pattern recognition receptors (PRRs) are involved in the pathogenesis of diseases, including Toll-like receptors (TLRs), C-type lectin receptors (CLRs), and Nod-like receptors (NLRs). Patients with primary immune deficiencies (PIDs) affecting TLR signaling can elucidate the importance of these proteins in the human immune system. Defects in interleukin-1 receptor-associated kinase-4 and myeloid differentiation factor 88 (MyD88) lead to susceptibility to infections with bacteria, while mutations in nuclear factor-κB essential modulator (NEMO) and other downstream mediators generally induce broader susceptibility to bacteria, viruses, and fungi. In contrast, TLR3 signaling defects are associated with susceptibility to herpes simplex virus type 1 encephalitis. Other PIDs induce functional alterations of TLR signaling pathways, such as common variable immunodeficiency in which plasmacytoid dendritic cell defects enhance defective responses of B cells to shared TLR agonists. Altered TLR responses to TLR2 and 4 agonists are seen in chronic granulomatous disease (CGD) and X-linked agammaglobulinemia (XLA). Enhanced TLR responses, meanwhile, are seen for TLRs 5 and 9 in CGD, TLRs 4, 7/8, and 9 in XLA, TLRs 2 and 4 in hyper IgE syndrome (HIES), and for most TLRs in adenosine deaminase deficiency. In this review we provide the reader with an update on the role of TLRs and downstream signaling pathways in PID disorders.
Collapse
Affiliation(s)
- Esameil Mortaz
- Department of Immunology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ian M Adcock
- Airways Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Payam Tabarsi
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ilad Alavi Darazam
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti, University of Medical Sciences,Tehran, Iran
| | - Masoud Movassaghi
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles (UCLA), USA
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Sciences, Utrecht University, Utrecht, The Netherlands; Department of Immunology, Nutricia Research, Utrecht, the Netherlands
| | - Hamidreza Jamaati
- Chronic Respiratory Diseases Research Center and National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Aliakbar Velayati
- Mycobacteriology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Chorny A, Casas-Recasens S, Sintes J, Shan M, Polentarutti N, García-Escudero R, Walland AC, Yeiser JR, Cassis L, Carrillo J, Puga I, Cunha C, Bastos H, Rodrigues F, Lacerda JF, Morais A, Dieguez-Gonzalez R, Heeger PS, Salvatori G, Carvalho A, Garcia-Sastre A, Blander JM, Mantovani A, Garlanda C, Cerutti A. The soluble pattern recognition receptor PTX3 links humoral innate and adaptive immune responses by helping marginal zone B cells. J Exp Med 2016; 213:2167-85. [PMID: 27621420 PMCID: PMC5030794 DOI: 10.1084/jem.20150282] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 08/04/2016] [Indexed: 01/01/2023] Open
Abstract
Cerutti and collaborators show that the humoral arms of the innate and adaptive immune systems are functionally interconnected by pentraxin 3, a soluble pattern recognition receptor that couples innate immune recognition with antibody-inducing function. Pentraxin 3 (PTX3) is a fluid-phase pattern recognition receptor of the humoral innate immune system with ancestral antibody-like properties but unknown antibody-inducing function. In this study, we found binding of PTX3 to splenic marginal zone (MZ) B cells, an innate-like subset of antibody-producing lymphocytes strategically positioned at the interface between the circulation and the adaptive immune system. PTX3 was released by a subset of neutrophils that surrounded the splenic MZ and expressed an immune activation–related gene signature distinct from that of circulating neutrophils. Binding of PTX3 promoted homeostatic production of IgM and class-switched IgG antibodies to microbial capsular polysaccharides, which decreased in PTX3-deficient mice and humans. In addition, PTX3 increased IgM and IgG production after infection with blood-borne encapsulated bacteria or immunization with bacterial carbohydrates. This immunogenic effect stemmed from the activation of MZ B cells through a neutrophil-regulated pathway that elicited class switching and plasmablast expansion via a combination of T cell–independent and T cell–dependent signals. Thus, PTX3 may bridge the humoral arms of the innate and adaptive immune systems by serving as an endogenous adjuvant for MZ B cells. This property could be harnessed to develop more effective vaccines against encapsulated pathogens.
Collapse
Affiliation(s)
- Alejo Chorny
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Sandra Casas-Recasens
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Jordi Sintes
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques, 08003 Barcelona, Spain
| | - Meimei Shan
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Nadia Polentarutti
- Istituto di Ricovero e Cura a Carattere Scientifico Humanitas Clinical and Research Hospital, Rozzano, 20089 Milan, Italy
| | - Ramón García-Escudero
- Molecular Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales, y Tecnológicas, 28040 Madrid, Spain
| | - A Cooper Walland
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - John R Yeiser
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Linda Cassis
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques, 08003 Barcelona, Spain
| | - Jorge Carrillo
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 Institut de Recerca de la Sida IrsiCaixa, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Barcelona, Spain
| | - Irene Puga
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques, 08003 Barcelona, Spain
| | - Cristina Cunha
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, 4710-057 Braga, Portugal PT Government Associate Laboratory, Braga/Guimarães, Life and Health Sciences Research Institute /3B's, University of Minho, 4710-057 Braga, Portugal
| | - Hélder Bastos
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, 4710-057 Braga, Portugal PT Government Associate Laboratory, Braga/Guimarães, Life and Health Sciences Research Institute /3B's, University of Minho, 4710-057 Braga, Portugal Serviço de Pneumologia, Centro Hospitalar São João, Faculdade de Medicina, Universidade do Porto, 4200-319 Porto, Portugal
| | - Fernando Rodrigues
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, 4710-057 Braga, Portugal PT Government Associate Laboratory, Braga/Guimarães, Life and Health Sciences Research Institute /3B's, University of Minho, 4710-057 Braga, Portugal
| | - João F Lacerda
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal Hospital de Santa Maria, 1649-035 Lisboa, Portugal
| | - António Morais
- Serviço de Pneumologia, Centro Hospitalar São João, Faculdade de Medicina, Universidade do Porto, 4200-319 Porto, Portugal
| | - Rebeca Dieguez-Gonzalez
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Peter S Heeger
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 Translational Transplant Research Center and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | | | - Agostinho Carvalho
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, 4710-057 Braga, Portugal PT Government Associate Laboratory, Braga/Guimarães, Life and Health Sciences Research Institute /3B's, University of Minho, 4710-057 Braga, Portugal
| | - Adolfo Garcia-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - J Magarian Blander
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Alberto Mantovani
- Istituto di Ricovero e Cura a Carattere Scientifico Humanitas Clinical and Research Hospital, Rozzano, 20089 Milan, Italy Humanitas University, Rozzano, 20089 Milan, Italy
| | - Cecilia Garlanda
- Istituto di Ricovero e Cura a Carattere Scientifico Humanitas Clinical and Research Hospital, Rozzano, 20089 Milan, Italy Humanitas University, Rozzano, 20089 Milan, Italy
| | - Andrea Cerutti
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques, 08003 Barcelona, Spain Catalan Institute for Research and Advanced Studies, 08003 Barcelona, Spain
| |
Collapse
|
16
|
Analysis of TLR4 (Asp299Gly and Thr399Ile) gene polymorphisms and mRNA level in patients with dengue infection: A case-control study. INFECTION GENETICS AND EVOLUTION 2016; 43:412-7. [DOI: 10.1016/j.meegid.2016.06.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 06/07/2016] [Accepted: 06/10/2016] [Indexed: 12/17/2022]
|
17
|
Rack-Hoch A, Notheis G, Klein C, Hauck F. [Basic diagnostics for pediatric outpatients with susceptibility to infections and immundysregulation]. MMW Fortschr Med 2016; 158:49-52. [PMID: 27221434 DOI: 10.1007/s15006-016-8280-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Affiliation(s)
- Anita Rack-Hoch
- Immundefektambulanz, Dr. von Haunersches Kinderspital, Ludwig-Maximilians-Universität München, Lindwurmstr. 4, D-80999, München, Deutschland.
| | - Gundula Notheis
- Immundefektambulanz, Dr. von Haunersches Kinderspital, Ludwig-Maximilians-Universität München, Lindwurmstr. 4, D-80999, München, Deutschland
| | - Christoph Klein
- Immundefektambulanz, Dr. von Haunersches Kinderspital, Ludwig-Maximilians-Universität München, Lindwurmstr. 4, D-80999, München, Deutschland
| | - Fabian Hauck
- Immundefektambulanz, Dr. von Haunersches Kinderspital, Ludwig-Maximilians-Universität München, Lindwurmstr. 4, D-80999, München, Deutschland
| |
Collapse
|
18
|
Cerwenka A, Lanier LL. Natural killer cell memory in infection, inflammation and cancer. Nat Rev Immunol 2016; 16:112-23. [PMID: 26806484 DOI: 10.1038/nri.2015.9] [Citation(s) in RCA: 386] [Impact Index Per Article: 48.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Immunological memory can be defined as a quantitatively and qualitatively enhanced immune response upon rechallenge. For natural killer (NK) cells, two main types of memory exist. First, similarly to T cells and B cells, NK cells can exert immunological memory after encounters with stimuli such as haptens or viruses, resulting in the generation of antigen-specific memory NK cells. Second, NK cells can remember inflammatory cytokine milieus that imprint long-lasting non-antigen-specific NK cell effector function. The basic concepts derived from studying NK cell memory provide new insights about innate immunity and could lead to novel strategies to improve treatments for infectious diseases and cancer.
Collapse
Affiliation(s)
- Adelheid Cerwenka
- Innate Immunity Group, German Cancer Research Center/D080, 69120 Heidelberg, Germany
| | - Lewis L Lanier
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, California 94143, USA
| |
Collapse
|
19
|
Kim Y, Jeong EJ, Han Lee IS, Kim MY, Cho JY. (E)-3-(3-methoxyphenyl)-1-(2-pyrrolyl)-2-propenone displays suppression of inflammatory responses via inhibition of Src, Syk, and NF-κB. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2015; 20:91-9. [PMID: 26807028 PMCID: PMC4722197 DOI: 10.4196/kjpp.2016.20.1.91] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Revised: 11/05/2015] [Accepted: 11/10/2015] [Indexed: 12/24/2022]
Abstract
(E)-3-(3-methoxyphenyl)-1-(2-pyrrolyl)-2-propenone (MPP) is an aldol condensation product resulting from pyrrole-2-carbaldehyde and m- and p- substituted acetophenones. However, its biological activity has not yet been evaluated. Since it has been reported that some propenone-type compounds display anti-inflammatory activity, we investigated whether MPP could negatively modulate inflammatory responses. To do this, we employed lipopolysaccharide (LPS)-stimulated macrophage-like RAW264.7 cells and examined the inhibitory levels of nitric oxide (NO) production and transcriptional activation, as well as the target proteins involved in the inflammatory signaling cascade. Interestingly, MPP was found to reduce the production of NO in LPS-treated RAW264.7 cells, without causing cytotoxicity. Moreover, this compound suppressed the mRNA levels of inflammatory genes, such as inducible NO synthase (iNOS) and tumor necrosis factor (TNF)-α. Using luciferase reporter gene assays performed in HEK293 cells and immunoblotting analysis with nuclear protein fractions, we determined that MPP reduced the transcriptional activation of nuclear factor (NF)-κB. Furthermore, the activation of a series of upstream signals for NF-κB activation, composed of Src, Syk, Akt, and IκBα, were also blocked by this compound. It was confirmed that MPP was able to suppress autophosphorylation of overexpressed Src and Syk in HEK293 cells. Therefore, these results suggest that MPP can function as an anti-inflammatory drug with NF-κB inhibitory properties via the suppression of Src and Syk.
Collapse
Affiliation(s)
- Yong Kim
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Korea
| | - Eun Jeong Jeong
- Department of Science Education, Kangwon National University, Chuncheon 24341, Korea
| | - In-Sook Han Lee
- Department of Science Education, Kangwon National University, Chuncheon 24341, Korea
| | - Mi-Yeon Kim
- Department of Bioinformatics and Life Science, Soongsil University, Seoul 06978, Korea
| | - Jae Youl Cho
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Korea
| |
Collapse
|
20
|
Tan KW, Griffiths CEM. Novel systemic therapies for the treatment of psoriasis. Expert Opin Pharmacother 2015; 17:79-92. [DOI: 10.1517/14656566.2016.1109636] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
21
|
Antonietti J, Retornaz K, Bernasconi A, Laporte RJ, Minodier P, Bustamante JC, Dubus JC. [Disseminated BCG disease revealing a partial deficiency in receptor 1 interferon gamma]. Arch Pediatr 2015; 22:971-3. [PMID: 26251056 DOI: 10.1016/j.arcped.2015.06.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 06/18/2015] [Accepted: 06/19/2015] [Indexed: 11/19/2022]
Abstract
We report on a case of disseminated BCGitis with an unusual presentation in a 4-month-old infant revealing a syndrome of Mendelian susceptibility to mycobacteria due to a partial dominant mutation of the interferon gamma receptor 1 gene.
Collapse
Affiliation(s)
- J Antonietti
- Service de médecine infantile, hôpital Nord, chemin des Bourrely, 13015 Marseille, France.
| | - K Retornaz
- Service de médecine infantile, hôpital Nord, chemin des Bourrely, 13015 Marseille, France
| | - A Bernasconi
- Service de médecine infantile, hôpital Nord, chemin des Bourrely, 13015 Marseille, France
| | - R-J Laporte
- Service d'accueil des urgences pédiatriques, hôpital Nord, chemin des Bourrely, 13015 Marseille, France
| | - P Minodier
- Service d'accueil des urgences pédiatriques, hôpital Nord, chemin des Bourrely, 13015 Marseille, France
| | - J-C Bustamante
- Laboratoire de génétique humaine des maladies infectieuses, institut Imagine, 24, boulevard de Montparnasse, 75015 Paris, France
| | - J-C Dubus
- Service de médecine infantile, hôpital Nord, chemin des Bourrely, 13015 Marseille, France; Service d'accueil des urgences pédiatriques, hôpital Nord, chemin des Bourrely, 13015 Marseille, France
| |
Collapse
|
22
|
IL-23/IL-17A Dysfunction Phenotypes Inform Possible Clinical Effects from Anti-IL-17A Therapies. J Invest Dermatol 2015; 135:1946-1953. [PMID: 25972190 PMCID: PMC4580732 DOI: 10.1038/jid.2015.144] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 03/10/2015] [Accepted: 03/24/2015] [Indexed: 12/14/2022]
Abstract
Biologics that neutralize specific cytokines have improved outcomes for several immune-mediated disorders but may also increase risks for particular side effects. This article postulates potential immunologic consequences of inhibiting components of the IL-23/T-helper cell 17 pathway–the target of next-generation biologics for treating psoriasis–based on clinical phenotypes of inherent or acquired deficiencies in this pathway. Generally, downstream deficiencies (e.g., IL-17A, IL-17F) are associated with fewer disorders compared with upstream deficiencies, suggesting that selectively blocking downstream targets may result in a narrower range of side effects. However, safety of these specific inhibitions must be established in long-term studies.
Collapse
|
23
|
Rare mendelian primary immunodeficiency diseases associated with impaired NF-κB signaling. Genes Immun 2015; 16:239-46. [PMID: 25764117 PMCID: PMC4457537 DOI: 10.1038/gene.2015.3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 01/07/2015] [Accepted: 01/09/2015] [Indexed: 12/14/2022]
Abstract
Mendelian Primary Immunodeficiency Diseases (MPIDs) are rare disorders affecting distinct constituents of the innate and adaptive immune system. Although they are genetically heterogeneous a substantial group of MPIDs is due to mutations in genes affecting the NF-κB transcription pathway, essential for cell proliferation, cell survival, and involved in innate immunity and in inflammation. Many of these genes encode for crucial regulatory components of NF-κB pathway and their mutations are associated with immunological and developmental signs somehow overlapping in patients with MPIDs. At present nine different MPIDs listed in the OMIM are caused by mutations in at least nine different genes strictly involved in the NF-κB pathway that result in defects in immune responses. We will report here on the distinct function of each causative gene, on the impaired NF-κB step and more in general on the molecular mechanisms underlining the pathogenesis of the disease. Overall, the MPIDs affecting NF-κB signalosome require a careful integrated diagnosis and appropriate genetic tests to be molecularly identified. Their discovery at an ever-increasing rate will help to establish common therapeutic strategy for a subclass of immunodeficient patients.
Collapse
|
24
|
MacLennan CA, Martin LB, Micoli F. Vaccines against invasive Salmonella disease: current status and future directions. Hum Vaccin Immunother 2014; 10:1478-93. [PMID: 24804797 PMCID: PMC4185946 DOI: 10.4161/hv.29054] [Citation(s) in RCA: 150] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Though primarily enteric pathogens, Salmonellae are responsible for a considerable yet under-appreciated global burden of invasive disease. In South and South-East Asia, this manifests as enteric fever caused by serovars Typhi and Paratyphi A. In sub-Saharan Africa, a similar disease burden results from invasive nontyphoidal Salmonellae, principally serovars Typhimurium and Enteritidis. The existing Ty21a live-attenuated and Vi capsular polysaccharide vaccines target S. Typhi and are not effective in young children where the burden of invasive Salmonella disease is highest. After years of lack of investment in new Salmonella vaccines, recent times have seen increased interest in the area led by emerging-market manufacturers, global health vaccine institutes and academic partners. New glycoconjugate vaccines against S. Typhi are becoming available with similar vaccines against other invasive serovars in development. With other new vaccines under investigation, including live-attenuated, protein-based and GMMA vaccines, now is an exciting time for the Salmonella vaccine field.
Collapse
Affiliation(s)
- Calman A MacLennan
- Novartis Vaccines Institute for Global Health; Siena, Italy; Medical Research Council Centre for Immune Regulation and Clinical Immunology Service; Institute of Biomedical Research, School of Immunity and Infection; College of Medicine and Dental Sciences; University of Birmingham; Birmingham, UK
| | - Laura B Martin
- Novartis Vaccines Institute for Global Health; Siena, Italy
| | | |
Collapse
|
25
|
Stackaruk ML, Lee AJ, Ashkar AA. Type I interferon regulation of natural killer cell function in primary and secondary infections. Expert Rev Vaccines 2014; 12:875-84. [PMID: 23984959 DOI: 10.1586/14760584.2013.814871] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The priming of natural killer (NK) cells by type I interferon (IFN) is necessary for protection against primary and secondary viral infections. However, the pathway by which type I IFN activates NK cells to elicit antiviral responses is controversial. There is evidence to suggest that type I IFN priming of NK cells occurs through both direct and indirect pathways. As with many innate mechanisms, type I IFN and NK cells also orchestrate the adaptive immune response and thus aid in protection against secondary infections. Type I IFN can shape CD4(+) T cell, B cell and humoral memory formation. In addition, long-lived NK cells can perform specific and enhanced memory-like protection in secondary infections. This review outlines the different mechanisms underlying type I IFN regulation of NK cells and how type I IFN and NK cells can be used as a therapeutic target in vaccinations.
Collapse
Affiliation(s)
- Michele L Stackaruk
- Department of Pathology and Molecular Medicine, Institute for Infectious Disease Research, McMaster Immunology Research Centre, McMaster University, Hamilton, MDCL 4015, 1280 Main Street West, Hamilton, L8S 4K1, ON, Canada
| | | | | |
Collapse
|
26
|
Siggins MK, O'Shaughnessy CM, Pravin J, Cunningham AF, Henderson IR, Drayson MT, MacLennan CA. Differential timing of antibody-mediated phagocytosis and cell-free killing of invasive African Salmonella allows immune evasion. Eur J Immunol 2014; 44:1093-8. [PMID: 24375424 DOI: 10.1002/eji.201343529] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2013] [Revised: 10/17/2013] [Accepted: 12/19/2013] [Indexed: 11/07/2022]
Abstract
Nontyphoidal Salmonellae commonly cause fatal bacteraemia in African children lacking anti-Salmonella antibodies. These are facultative intracellular bacteria capable of cell-free and intracellular survival within macrophages. To better understand the relationship between extracellular and intracellular infection in blood and general mechanisms of Ab-related protection against Salmonella, we used human blood and sera to measure kinetics of Ab and complement deposition, serum-mediated bactericidal killing and phagocytosis of invasive African Salmonella enterica serovar Typhimurium D23580. Binding of antibodies peaked by 30 s, but C3 deposition lagged behind, peaking after 2-4 min. C5b-9 deposition was undetectable until between 2 and 6 min and peaked after 10 min, after which time an increase in serum-mediated killing occurred. In contrast, intracellular, opsonized Salmonellae were readily detectable within 5 min. By 10 min, around half of monocytes and most neutrophils contained bacteria. The same kinetics of serum-mediated killing and phagocytosis were observed with S. enterica Typhimurium laboratory strain SL1344, and the S. enterica Enteritidis African invasive isolate D24954 and laboratory strain PT4. The differential kinetics between cell-free killing and phagocytosis of invasive nontyphoidal Salmonella allows these bacteria to escape the blood and establish intracellular infection before they are killed by the membrane attack complex.
Collapse
Affiliation(s)
- Matthew K Siggins
- Medical Research Council Centre for Immune Regulation and Clinical Immunology Service, Institute of Biomedical Research, School of Immunity and Infection, College of Medicine and Dental Sciences, University of Birmingham, Birmingham, UK; Novartis Vaccines Institute for Global Health, Siena, Italy
| | | | | | | | | | | | | |
Collapse
|
27
|
Laws PM, Warren RB. Ustekinumab for the treatment of psoriasis. Expert Rev Clin Immunol 2014; 7:155-64. [DOI: 10.1586/eci.11.4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
28
|
Vergou T, Vasconcelos Lima XT, Kimball AB. Targeting the IL-12/IL-23 cytokine family in the treatment of psoriatic disease. ACTA ACUST UNITED AC 2014. [DOI: 10.1586/17469872.3.4.453] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
29
|
Nontuberculous mycobacterial infections in children with inborn errors of the immune system. J Infect 2013; 68 Suppl 1:S134-50. [PMID: 24119826 DOI: 10.1016/j.jinf.2013.09.024] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2013] [Indexed: 11/22/2022]
Abstract
Severe mycobacterial disease is mostly confined to patients who are immunocompromized either by acquired or inherited causes. One such genetic disorder is Mendelian Susceptibility to Mycobacterial Disease (MSMD), a hot topic within the field of primary immunodeficiency. This single gene disorder is characterized by isolated infection with mycobacteria or Salmonella due to a defect in the type-1 cytokine response. In the last two decades, ten genes have been labeled as causing MSMD when they harbor germline mutations, namely IL12B, IL12RB1, IFNGR1, IFNGR2, STAT1, IKBKG, CYBB, TYK2, IRF8 and ISG15. The mutations lead to either insufficient production of IFN-γ, or to an insufficient response to the cytokine. Current treatment options include recombinant IFN-γ and hematologic stem cell transplantation (HSCT). In the future, gene therapy, antisense-mediated exon skipping and chemical intervention in glycosylation problems may become successful alternatives. Furthermore, it is likely that many new candidate genes and pathways crucial for mycobacterial immunity will be identified.
Collapse
|
30
|
Fowler CJ, Olivier KN, Leung JM, Smith CC, Huth AG, Root H, Kuhns DB, Logun C, Zelazny A, Frein CA, Daub J, Haney C, Shelhamer JH, Bryant CE, Holland SM. Abnormal nasal nitric oxide production, ciliary beat frequency, and Toll-like receptor response in pulmonary nontuberculous mycobacterial disease epithelium. Am J Respir Crit Care Med 2013; 187:1374-81. [PMID: 23593951 DOI: 10.1164/rccm.201212-2197oc] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Pulmonary nontuberculous mycobacterial (PNTM) disease has increased over the past several decades, especially in older women. Despite extensive investigation, no consistent immunological abnormalities have been found. Using evidence from diseases such as cystic fibrosis and primary ciliary dyskinesia, in which mucociliary dysfunction predisposes subjects to high rates of nontuberculous mycobacterial disease that increase with age, we investigated correlates of mucociliary function in subjects with PNTM infections and healthy control subjects. OBJECTIVES To define ex vivo characteristics of PNTM disease. METHODS From 2009 to 2012, 58 subjects with PNTM infections and 40 control subjects were recruited. Nasal nitric oxide (nNO) was determined at the time of respiratory epithelial collection. Ciliary beat frequency at rest and in response to Toll-like receptor (TLR) and other agonists was determined using high-speed video microscopy. MEASUREMENTS AND MAIN RESULTS We found decreased nNO production, abnormally low resting ciliary beat frequency, and abnormal responses to agonists of TLR2, -3, -5, -7/8, and -9 in subjects with PNTM compared with healthy control subjects. The low ciliary beat frequency in subjects with PNTM was normalized ex vivo by augmentation of the NO-cyclic guanosine monophosphate pathway without normalization of their TLR agonist responses. CONCLUSIONS Impaired nNO, ciliary beat frequency, and TLR responses in PNTM disease epithelium identify possible underlying susceptibility mechanisms as well as possible avenues for directed investigation and therapy.
Collapse
Affiliation(s)
- Cedar J Fowler
- Immunopathogenesis Section, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892-1684, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
ISG15 is a well-known intracellular ubiquitin-like molecule involved in ISGylation. However, a recent study has revived the notion first put forward two decades ago that ISG15 is also a secreted molecule. Human neutrophils, monocytes and lymphocytes can release ISG15, even though this protein has no detectable signal peptide sequence. ISG15 has also been found in the secretory granules of granulocytes. The mechanism underlying ISG15 secretion is unknown. Secreted ISG15 acts on at least T and natural killer (NK) lymphocytes, in which it induces interferon (IFN)-γ production. However, the mechanism by which ISG15 stimulates these cells also remains unclear. ISG15 and IFN-γ seem to define an innate circuit that operates preferentially, but not exclusively, between granulocytes and NK cells. Inherited ISG15 deficiency is associated with severe mycobacterial disease in both mice and humans. This infectious phenotype probably results from the lack of secreted ISG15, because patients and mice with other inborn errors of IFN-γ immunity also display mycobacterial diseases. In addition to raising mechanistic issues, the studies described here pave the way for clinical studies of various aspects, ranging from the use of recombinant ISG15 in patients with infectious diseases to the use of ISG15-blocking agents in patients with inflammatory diseases.
Collapse
|
32
|
Picard C. [When should immunologic explorations be carried out in children with suspicion of primary immunodeficiency?]. Arch Pediatr 2013; 20:412-7. [PMID: 23480855 DOI: 10.1016/j.arcped.2013.01.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 01/07/2013] [Accepted: 01/19/2013] [Indexed: 11/29/2022]
Abstract
The identification of primary immunodeficiency is important in the prognosis and treatment of patients with these rare genetic diseases. For their diagnosis, simple screening explorations need to be carried out in case of severe, recurrent, and/or unusual infections. These include the whole blood cell count, plasmatic immunoglobulin levels, and postimmunization and/or post-infectious serologies. These examinations are used to guide the diagnosis with the joint analysis of patient's medical history, the clinical examination, and screening of biological results. They will then guide the prescription of more specific second-line explorations depending on the type of primary immunodeficiency suspected.
Collapse
Affiliation(s)
- C Picard
- Centre d'étude des déficits immunitaires (CEDI), pavillon Kirmisson, hôpital Necker-Enfants Malades, AP-HP, 149, rue de Sèvres, 75015 Paris, France.
| |
Collapse
|
33
|
Kong XF, Bousfiha A, Rouissi A, Itan Y, Abhyankar A, Bryant V, Okada S, Ailal F, Bustamante J, Casanova JL, Hirst J, Boisson-Dupuis S. A novel homozygous p.R1105X mutation of the AP4E1 gene in twins with hereditary spastic paraplegia and mycobacterial disease. PLoS One 2013; 8:e58286. [PMID: 23472171 PMCID: PMC3589270 DOI: 10.1371/journal.pone.0058286] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Accepted: 02/01/2013] [Indexed: 12/22/2022] Open
Abstract
We report identical twins with intellectual disability, progressive spastic paraplegia and short stature, born to a consanguineous family. Intriguingly, both children presented with lymphadenitis caused by the live Bacillus Calmette-Guérin (BCG) vaccine. Two syndromes – hereditary spastic paraplegia (HSP) and mycobacterial disease – thus occurred simultaneously. Whole-exome sequencing (WES) revealed a homozygous nonsense mutation (p.R1105X) of the AP4E1 gene, which was confirmed by Sanger sequencing. The p.R1105X mutation has no effect on AP4E1 mRNA levels, but results in lower levels of AP-4ε protein and of the other components of the AP-4 complex, as shown by western blotting, immunoprecipitation and immunofluorescence. Thus, the C-terminal part of the AP-4ε subunit plays an important role in maintaining the integrity of the AP-4 complex. No abnormalities of the IL-12/IFN-γ axis or oxidative burst pathways were identified. In conclusion, we identified twins with autosomal recessive AP-4 deficiency associated with HSP and mycobacterial disease, suggesting that AP-4 may play important role in the neurological and immunological systems.
Collapse
Affiliation(s)
- Xiao-Fei Kong
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Prando C, Samarina A, Bustamante J, Boisson-Dupuis S, Cobat A, Picard C, AlSum Z, Al-Jumaah S, Al-Hajjar S, Frayha H, Al-Mousa H, Ben-Mustapha I, Adimi P, Feinberg J, de Suremain M, Jannière L, Filipe-Santos O, Mansouri N, Stephan JL, Nallusamy R, Kumararatne DS, Bloorsaz MR, Ben-Ali M, Elloumi-Zghal H, Chemli J, Bouguila J, Bejaoui M, Alaki E, AlFawaz TS, Al Idrissi E, ElGhazali G, Pollard AJ, Murugasu B, Wah Lee B, Halwani R, Al-Zahrani M, Al Shehri MA, Al-Zahrani M, Bin-Hussain I, Mahdaviani SA, Parvaneh N, Abel L, Mansouri D, Barbouche R, Al-Muhsen S, Casanova JL. Inherited IL-12p40 deficiency: genetic, immunologic, and clinical features of 49 patients from 30 kindreds. Medicine (Baltimore) 2013; 92:109-122. [PMID: 23429356 PMCID: PMC3822760 DOI: 10.1097/md.0b013e31828a01f9] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Autosomal recessive interleukin (IL)-12 p40 (IL-12p40) deficiency is a rare genetic etiology of mendelian susceptibility to mycobacterial disease (MSMD). We report the genetic, immunologic, and clinical features of 49 patients from 30 kindreds originating from 5 countries (India, Iran, Pakistan, Saudi Arabia, and Tunisia). There are only 9 different mutant alleles of the IL12B gene: 2 small insertions, 3 small deletions, 2 splice site mutations, and 1 large deletion, each causing a frameshift and leading to a premature stop codon, and 1 nonsense mutation. Four of these 9 variants are recurrent, affecting 25 of the 30 reported kindreds, due to founder effects in specific countries. All patients are homozygous and display complete IL-12p40 deficiency. As a result, the patients lack detectable IL-12p70 and IL-12p40 and have low levels of interferon gamma (IFN-γ). The clinical features are characterized by childhood onset of bacille Calmette-Guérin (attenuated Mycobacterium bovis strain) (BCG) and Salmonella infections, with recurrences of salmonellosis (36.4%) more common than recurrences of mycobacterial disease (25%). BCG vaccination led to BCG disease in 40 of the 41 patients vaccinated (97.5%). Multiple mycobacterial infections were rare, observed in only 3 patients, whereas the association of salmonellosis and mycobacteriosis was observed in 9 patients. A few other infections were diagnosed, including chronic mucocutaneous candidiasis (n = 3), nocardiosis (n = 2), and klebsiellosis (n = 1). IL-12p40 deficiency has a high but incomplete clinical penetrance, with 33.3% of genetically affected relatives of index cases showing no symptoms. However, the prognosis is poor, with mortality rates of up to 28.6%. Overall, the clinical phenotype of IL-12p40 deficiency closely resembles that of interleukin 12 receptor β1 (IL-12Rβ1) deficiency. In conclusion, IL-12p40 deficiency is more common than initially thought and should be considered worldwide in patients with MSMD and other intramacrophagic infectious diseases, salmonellosis in particular.
Collapse
Affiliation(s)
| | - Arina Samarina
- From the St. Giles Laboratory of Human Genetics of Infectious Diseases (C. Prando, SBD, LA, JLC), Rockefeller Branch, The Rockefeller University, New York, New York; Laboratory of Human Genetics of Infectious Diseases (AS, J. Bustamante, SBD, C. Picard, JF, MdS, LJ, OFS, LA, JLC) Necker Branch, Institut National de la Santé et de la Recherche Médicale, U980, Necker Branch, Paris, France; University Paris Descartes (AS, J. Bustamante, SBD, C. Picard, JF, MdS, LJ, OFS, JLC) Paris Cité Sorbonne, Necker Medical School, Paris, France; Center for the Study of Primary Immunodeficiencies (J. Bustamante, C. Picard) and Pediatric Hematology-Immunology Unit (C. Picard, JLC), Assistance Publique-Hôpitaux de Paris, Necker Hospital, Paris, France; McGill Centre for the Study of Host Resistance (AC), Research Institute of McGill University Health Centre, and Departments of Human Genetics and Medicine, McGill University, Montreal, Quebec, Canada; Prince Naif Center for Immunology Research (ZAS, RH, S. Al-Muhsen, JLC) and Department of Pediatrics (ZAS, S. Al-Muhsen), College of Medicine, KingSaud University, Riyadh, Saudi Arabia; Department of Pediatrics (SAJ, SAH, HF, HAM, Mofareh Al-Zahrani, S. Al-Muhsen, IBH) King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia; King Saud Medical City (EA), Riyadh, Saudi Arabia; Laboratory of Cytoimmunology (IBM, MBA, HEZ, RB), Pasteur Institut of Tunis, Tunis-Belvédère, Tunisia; Department of Clinical Immunology and Infectious Disease (PA, NM, DM) and Pediatric Respiratory Disease Research Center (MRB S.A Mahdaviani), National Research Institute of Tuberculosis and Lung Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Pediatrics (JLS), University of Saint Etienne, Hôpital Nord, Saint Etienne, France; Department of Pediatrics (RN), Penang Medical College, Penang, Malaysia; Department of Clinical Biochemistry and Immunology (DSK), Addenbrookes Hospital, Cambridge, United Kingdom; Department of Pediatrics (JC), Sahloul Hospital, Sousse, Tunisia; Department of Pediatrics (J. Bouguila), Farhat Hached Hospital, Sousse, Tunisia; Department of Pediatrics (MB), Bone Marrow Transplantation Center, Tunis, Tunisia; Department of Pediatrics (TSAF, EAI, GEG, MAAS, Mofareh Al-Zahrani), King Fahad Medical City, Riyadh, Saudi Arabia; Department of Paediatrics (AJP), University of Oxford, NIHR Oxford Biomedical Research Centre, Children’s Hospital, Oxford, United Kingdom; Department of Pediatrics (BM, BWL), National University of Singapore, Singapore; Department of Pediatrics (Mohammed Al-Zahrani), Security Forces Hospital, Riyadh, Saudi Arabia; and Pediatric Infectious Disease Research Center (NP), Tehran University of Medical Sciences, Tehran, Iran
| | - Jacinta Bustamante
- From the St. Giles Laboratory of Human Genetics of Infectious Diseases (C. Prando, SBD, LA, JLC), Rockefeller Branch, The Rockefeller University, New York, New York; Laboratory of Human Genetics of Infectious Diseases (AS, J. Bustamante, SBD, C. Picard, JF, MdS, LJ, OFS, LA, JLC) Necker Branch, Institut National de la Santé et de la Recherche Médicale, U980, Necker Branch, Paris, France; University Paris Descartes (AS, J. Bustamante, SBD, C. Picard, JF, MdS, LJ, OFS, JLC) Paris Cité Sorbonne, Necker Medical School, Paris, France; Center for the Study of Primary Immunodeficiencies (J. Bustamante, C. Picard) and Pediatric Hematology-Immunology Unit (C. Picard, JLC), Assistance Publique-Hôpitaux de Paris, Necker Hospital, Paris, France; McGill Centre for the Study of Host Resistance (AC), Research Institute of McGill University Health Centre, and Departments of Human Genetics and Medicine, McGill University, Montreal, Quebec, Canada; Prince Naif Center for Immunology Research (ZAS, RH, S. Al-Muhsen, JLC) and Department of Pediatrics (ZAS, S. Al-Muhsen), College of Medicine, KingSaud University, Riyadh, Saudi Arabia; Department of Pediatrics (SAJ, SAH, HF, HAM, Mofareh Al-Zahrani, S. Al-Muhsen, IBH) King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia; King Saud Medical City (EA), Riyadh, Saudi Arabia; Laboratory of Cytoimmunology (IBM, MBA, HEZ, RB), Pasteur Institut of Tunis, Tunis-Belvédère, Tunisia; Department of Clinical Immunology and Infectious Disease (PA, NM, DM) and Pediatric Respiratory Disease Research Center (MRB S.A Mahdaviani), National Research Institute of Tuberculosis and Lung Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Pediatrics (JLS), University of Saint Etienne, Hôpital Nord, Saint Etienne, France; Department of Pediatrics (RN), Penang Medical College, Penang, Malaysia; Department of Clinical Biochemistry and Immunology (DSK), Addenbrookes Hospital, Cambridge, United Kingdom; Department of Pediatrics (JC), Sahloul Hospital, Sousse, Tunisia; Department of Pediatrics (J. Bouguila), Farhat Hached Hospital, Sousse, Tunisia; Department of Pediatrics (MB), Bone Marrow Transplantation Center, Tunis, Tunisia; Department of Pediatrics (TSAF, EAI, GEG, MAAS, Mofareh Al-Zahrani), King Fahad Medical City, Riyadh, Saudi Arabia; Department of Paediatrics (AJP), University of Oxford, NIHR Oxford Biomedical Research Centre, Children’s Hospital, Oxford, United Kingdom; Department of Pediatrics (BM, BWL), National University of Singapore, Singapore; Department of Pediatrics (Mohammed Al-Zahrani), Security Forces Hospital, Riyadh, Saudi Arabia; and Pediatric Infectious Disease Research Center (NP), Tehran University of Medical Sciences, Tehran, Iran
| | - Stéphanie Boisson-Dupuis
- From the St. Giles Laboratory of Human Genetics of Infectious Diseases (C. Prando, SBD, LA, JLC), Rockefeller Branch, The Rockefeller University, New York, New York; Laboratory of Human Genetics of Infectious Diseases (AS, J. Bustamante, SBD, C. Picard, JF, MdS, LJ, OFS, LA, JLC) Necker Branch, Institut National de la Santé et de la Recherche Médicale, U980, Necker Branch, Paris, France; University Paris Descartes (AS, J. Bustamante, SBD, C. Picard, JF, MdS, LJ, OFS, JLC) Paris Cité Sorbonne, Necker Medical School, Paris, France; Center for the Study of Primary Immunodeficiencies (J. Bustamante, C. Picard) and Pediatric Hematology-Immunology Unit (C. Picard, JLC), Assistance Publique-Hôpitaux de Paris, Necker Hospital, Paris, France; McGill Centre for the Study of Host Resistance (AC), Research Institute of McGill University Health Centre, and Departments of Human Genetics and Medicine, McGill University, Montreal, Quebec, Canada; Prince Naif Center for Immunology Research (ZAS, RH, S. Al-Muhsen, JLC) and Department of Pediatrics (ZAS, S. Al-Muhsen), College of Medicine, KingSaud University, Riyadh, Saudi Arabia; Department of Pediatrics (SAJ, SAH, HF, HAM, Mofareh Al-Zahrani, S. Al-Muhsen, IBH) King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia; King Saud Medical City (EA), Riyadh, Saudi Arabia; Laboratory of Cytoimmunology (IBM, MBA, HEZ, RB), Pasteur Institut of Tunis, Tunis-Belvédère, Tunisia; Department of Clinical Immunology and Infectious Disease (PA, NM, DM) and Pediatric Respiratory Disease Research Center (MRB S.A Mahdaviani), National Research Institute of Tuberculosis and Lung Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Pediatrics (JLS), University of Saint Etienne, Hôpital Nord, Saint Etienne, France; Department of Pediatrics (RN), Penang Medical College, Penang, Malaysia; Department of Clinical Biochemistry and Immunology (DSK), Addenbrookes Hospital, Cambridge, United Kingdom; Department of Pediatrics (JC), Sahloul Hospital, Sousse, Tunisia; Department of Pediatrics (J. Bouguila), Farhat Hached Hospital, Sousse, Tunisia; Department of Pediatrics (MB), Bone Marrow Transplantation Center, Tunis, Tunisia; Department of Pediatrics (TSAF, EAI, GEG, MAAS, Mofareh Al-Zahrani), King Fahad Medical City, Riyadh, Saudi Arabia; Department of Paediatrics (AJP), University of Oxford, NIHR Oxford Biomedical Research Centre, Children’s Hospital, Oxford, United Kingdom; Department of Pediatrics (BM, BWL), National University of Singapore, Singapore; Department of Pediatrics (Mohammed Al-Zahrani), Security Forces Hospital, Riyadh, Saudi Arabia; and Pediatric Infectious Disease Research Center (NP), Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Saleh Al-Muhsen
- From the St. Giles Laboratory of Human Genetics of Infectious Diseases (C. Prando, SBD, LA, JLC), Rockefeller Branch, The Rockefeller University, New York, New York; Laboratory of Human Genetics of Infectious Diseases (AS, J. Bustamante, SBD, C. Picard, JF, MdS, LJ, OFS, LA, JLC) Necker Branch, Institut National de la Santé et de la Recherche Médicale, U980, Necker Branch, Paris, France; University Paris Descartes (AS, J. Bustamante, SBD, C. Picard, JF, MdS, LJ, OFS, JLC) Paris Cité Sorbonne, Necker Medical School, Paris, France; Center for the Study of Primary Immunodeficiencies (J. Bustamante, C. Picard) and Pediatric Hematology-Immunology Unit (C. Picard, JLC), Assistance Publique-Hôpitaux de Paris, Necker Hospital, Paris, France; McGill Centre for the Study of Host Resistance (AC), Research Institute of McGill University Health Centre, and Departments of Human Genetics and Medicine, McGill University, Montreal, Quebec, Canada; Prince Naif Center for Immunology Research (ZAS, RH, S. Al-Muhsen, JLC) and Department of Pediatrics (ZAS, S. Al-Muhsen), College of Medicine, KingSaud University, Riyadh, Saudi Arabia; Department of Pediatrics (SAJ, SAH, HF, HAM, Mofareh Al-Zahrani, S. Al-Muhsen, IBH) King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia; King Saud Medical City (EA), Riyadh, Saudi Arabia; Laboratory of Cytoimmunology (IBM, MBA, HEZ, RB), Pasteur Institut of Tunis, Tunis-Belvédère, Tunisia; Department of Clinical Immunology and Infectious Disease (PA, NM, DM) and Pediatric Respiratory Disease Research Center (MRB S.A Mahdaviani), National Research Institute of Tuberculosis and Lung Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Pediatrics (JLS), University of Saint Etienne, Hôpital Nord, Saint Etienne, France; Department of Pediatrics (RN), Penang Medical College, Penang, Malaysia; Department of Clinical Biochemistry and Immunology (DSK), Addenbrookes Hospital, Cambridge, United Kingdom; Department of Pediatrics (JC), Sahloul Hospital, Sousse, Tunisia; Department of Pediatrics (J. Bouguila), Farhat Hached Hospital, Sousse, Tunisia; Department of Pediatrics (MB), Bone Marrow Transplantation Center, Tunis, Tunisia; Department of Pediatrics (TSAF, EAI, GEG, MAAS, Mofareh Al-Zahrani), King Fahad Medical City, Riyadh, Saudi Arabia; Department of Paediatrics (AJP), University of Oxford, NIHR Oxford Biomedical Research Centre, Children’s Hospital, Oxford, United Kingdom; Department of Pediatrics (BM, BWL), National University of Singapore, Singapore; Department of Pediatrics (Mohammed Al-Zahrani), Security Forces Hospital, Riyadh, Saudi Arabia; and Pediatric Infectious Disease Research Center (NP), Tehran University of Medical Sciences, Tehran, Iran
| | - Jean-Laurent Casanova
- From the St. Giles Laboratory of Human Genetics of Infectious Diseases (C. Prando, SBD, LA, JLC), Rockefeller Branch, The Rockefeller University, New York, New York; Laboratory of Human Genetics of Infectious Diseases (AS, J. Bustamante, SBD, C. Picard, JF, MdS, LJ, OFS, LA, JLC) Necker Branch, Institut National de la Santé et de la Recherche Médicale, U980, Necker Branch, Paris, France; University Paris Descartes (AS, J. Bustamante, SBD, C. Picard, JF, MdS, LJ, OFS, JLC) Paris Cité Sorbonne, Necker Medical School, Paris, France; Center for the Study of Primary Immunodeficiencies (J. Bustamante, C. Picard) and Pediatric Hematology-Immunology Unit (C. Picard, JLC), Assistance Publique-Hôpitaux de Paris, Necker Hospital, Paris, France; McGill Centre for the Study of Host Resistance (AC), Research Institute of McGill University Health Centre, and Departments of Human Genetics and Medicine, McGill University, Montreal, Quebec, Canada; Prince Naif Center for Immunology Research (ZAS, RH, S. Al-Muhsen, JLC) and Department of Pediatrics (ZAS, S. Al-Muhsen), College of Medicine, KingSaud University, Riyadh, Saudi Arabia; Department of Pediatrics (SAJ, SAH, HF, HAM, Mofareh Al-Zahrani, S. Al-Muhsen, IBH) King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia; King Saud Medical City (EA), Riyadh, Saudi Arabia; Laboratory of Cytoimmunology (IBM, MBA, HEZ, RB), Pasteur Institut of Tunis, Tunis-Belvédère, Tunisia; Department of Clinical Immunology and Infectious Disease (PA, NM, DM) and Pediatric Respiratory Disease Research Center (MRB S.A Mahdaviani), National Research Institute of Tuberculosis and Lung Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Pediatrics (JLS), University of Saint Etienne, Hôpital Nord, Saint Etienne, France; Department of Pediatrics (RN), Penang Medical College, Penang, Malaysia; Department of Clinical Biochemistry and Immunology (DSK), Addenbrookes Hospital, Cambridge, United Kingdom; Department of Pediatrics (JC), Sahloul Hospital, Sousse, Tunisia; Department of Pediatrics (J. Bouguila), Farhat Hached Hospital, Sousse, Tunisia; Department of Pediatrics (MB), Bone Marrow Transplantation Center, Tunis, Tunisia; Department of Pediatrics (TSAF, EAI, GEG, MAAS, Mofareh Al-Zahrani), King Fahad Medical City, Riyadh, Saudi Arabia; Department of Paediatrics (AJP), University of Oxford, NIHR Oxford Biomedical Research Centre, Children’s Hospital, Oxford, United Kingdom; Department of Pediatrics (BM, BWL), National University of Singapore, Singapore; Department of Pediatrics (Mohammed Al-Zahrani), Security Forces Hospital, Riyadh, Saudi Arabia; and Pediatric Infectious Disease Research Center (NP), Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
35
|
Yu T, Moh SH, Kim SB, Yang Y, Kim E, Lee YW, Cho CK, Kim KH, Yoo BC, Cho JY, Yoo HS. HangAmDan-B, an ethnomedicinal herbal mixture, suppresses inflammatory responses by inhibiting Syk/NF-κB and JNK/ATF-2 pathways. J Med Food 2012; 16:56-65. [PMID: 23256447 DOI: 10.1089/jmf.2012.2374] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
HangAmDan-B (HAD-B) is a powdered mixture of eight ethnopharmacologically characterized folk medicines that is prescribed for solid masses and cancers in Korea. In view of the finding that macrophage-mediated inflammation is a pathophysiologically important phenomenon, we investigated whether HAD-B modulates inflammatory responses and explored the associated molecular mechanisms. The immunomodulatory activity of HAD-B in toll-like receptor-activated macrophages induced by lipopolysaccharide (LPS) was assessed by measuring nitric oxide (NO) and prostaglandin E(2) (PGE(2)) levels. To identify the specific transcription factors (such as nuclear factor [NF]-κB and signaling enzymes) targeted by HAD-B, biochemical approaches, including kinase assays and immunoblot analysis, were additionally employed. HAD-B suppressed the production of PGE(2) and NO in LPS-activated macrophages in a dose-dependent manner. Furthermore, the extract ameliorated HCl/EtOH-induced gastritis symptoms. Moreover, HAD-B significantly inhibited LPS-induced mRNA expression of inducible NO synthase and cyclooxygenase (COX)-2. Interestingly, marked inhibition of NF-κB and activating transcription factor was observed in the presence of HAD-B. Data from direct kinase assays and immunoblot analysis showed that HAD-B suppresses activation of the upstream signaling cascade involving spleen tyrosine kinase, Src, p38, c-Jun N-terminal kinase, and transforming growth factor β-activated kinase 1. Finally, kaempferol, but not quercetin or resveratrol was identified as a bioactive compound in HAD-B. Therefore, our results suggest that HAD-B possesses anti-inflammatory activity that contributes to its anticancer property.
Collapse
Affiliation(s)
- Tao Yu
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Primary immunodeficiencies (PIDs) are severe defects in the capacity of the host to mount a proper immune response, and are characterized by an increased susceptibility to infections. Although classical immunodeficiencies have been characterized based on broad defects in cell populations (e.g. T/B cells or polymorphonuclear leukocytes) or humoral factors (e.g. antibodies or complement), specific immune defects based on well-defined molecular targets have been described more recently. Among these, genetic defects in pattern recognition receptors (PRRs), leading to impaired recognition of invading pathogens by the innate immune system, play an important role in specific defects against human pathogens. Defects have been described in three of the major families of PRRs: the Toll-like receptors, the C-type lectin receptors and the nucleotide-binding domain leucine-rich repeat-containing receptors. By contrast, no defects in the intracellular viral receptors of the RigI helicase family have been described to date. Defects in the PRRs show a broad variation in severity, have a narrow specificity for certain classes of pathogens, and often decrease in severity with age; these characteristics distinguish them from other forms of PIDs. Their discovery has led to important insights into the pathophysiology of infections, and may offer potential novel therapeutic targets for immunotherapy.
Collapse
Affiliation(s)
- M G Netea
- Department of Medicine and Nijmegen, Institute for Infection, Inflammation & Immunity (N4i), Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands.
| | | | | |
Collapse
|
37
|
Levy DE, Marié IJ, Durbin JE. Induction and function of type I and III interferon in response to viral infection. Curr Opin Virol 2012; 1:476-86. [PMID: 22323926 DOI: 10.1016/j.coviro.2011.11.001] [Citation(s) in RCA: 240] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The type I and III interferon (IFN) families consist of cytokines rapidly induced during viral infection that confer antiviral protection on target cells and are critical components of innate immune responses and the transition to effective adaptive immunity. The regulation of their expression involves an intricate and stringently regulated signaling cascade, initiated by recognition most often of viral nucleic acid in cytoplasmic and endosomal compartments and involving a series of protein conformational rearrangements and interactions regulated by helicase action, ubiquitin modification, and protein aggregation, culminating in kinase activation and phosphorylation of critical transcription factors and their regulators. The many IFN subtypes induced by viruses confer amplification, diversification, and cell-type specificity to the host response to infection, providing fertile ground for development of antiviral therapeutics and vaccines.
Collapse
Affiliation(s)
- David E Levy
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA.
| | | | | |
Collapse
|
38
|
KITAGISHI YASUKO, KOBAYASHI MAYUMI, YAMASHINA YURIE, MATSUDA SATORU. Elucidating the regulation of T cell subsets. Int J Mol Med 2012; 30:1255-60. [DOI: 10.3892/ijmm.2012.1152] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 09/19/2012] [Indexed: 11/05/2022] Open
|
39
|
Sharma AA, Jen R, Butler A, Lavoie PM. The developing human preterm neonatal immune system: a case for more research in this area. Clin Immunol 2012; 145:61-8. [PMID: 22926079 DOI: 10.1016/j.clim.2012.08.006] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Revised: 08/07/2012] [Accepted: 08/08/2012] [Indexed: 12/23/2022]
Abstract
Neonates, particularly those born prematurely, are among the most vulnerable age group for morbidity and mortality due to infections. Immaturity of the innate immune system and a high need for invasive medical procedures in the context of a preterm birth make these infants highly susceptible to common neonatal pathogens. Preterm infants who survive may also suffer permanent disabilities due to organ damage resulting from either the infection itself or from the inflammatory response generated under an oxidative stress. Infections in preterm infants continue to pose important healthcare challenges. Yet, developmental maturation events in the innate immune system that underlie their excessively high vulnerability to infection remain largely understudied. In this review article, we identify pertinent knowledge gaps that must be filled in order to orient future translational research.
Collapse
|
40
|
Kumar S, Ingle H, Prasad DVR, Kumar H. Recognition of bacterial infection by innate immune sensors. Crit Rev Microbiol 2012; 39:229-46. [DOI: 10.3109/1040841x.2012.706249] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
41
|
Mendelian susceptibility to mycobacterial disease in egyptian children. Mediterr J Hematol Infect Dis 2012; 4:e2012033. [PMID: 22708048 PMCID: PMC3375717 DOI: 10.4084/mjhid.2012.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Accepted: 04/13/2012] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Tuberculosis remains a major health problem in developing countries especially with the emergence of multidrug resistant strains. Mendelian Susceptibility to Mycobacterial Disease (MSMD) is a rare disorder with impaired immunity against mycobacterial pathogens. Reported MSMD etiologies highlight the crucial role of the Interferon gamma /Interleukin 12 (IFN-γ/ IL-12) axis and the phagocyte respiratory burst axis. PURPOSE Screen patients with possible presentations for MSMD. METHODS Patients with disseminated BCG infection following vaccination, atypical mycobacterial infections or recurrent tuberculosis infections were recruited from the Primary Immune Deficiency Clinic at Cairo University Specialized Pediatric Hospital, Egypt and immune and genetic laboratory investigations were conducted at Human Genetic of Infectious Diseases laboratory in Necker Medical School, France from 2005-2009. IFN-γ level in patient's plasma as well as mutations in the eight previously identified MSMD-causing genes were explored. RESULTS Nine cases from eight (unrelated) kindreds were evaluated in detail. We detected a high level of IFN-γ in plasma in one patient. Through Sanger sequencing, a homozygous mutation in the IFNGR1 gene at position 485 corresponding to an amino acid change from serine to phenylalanine (S485F), was detected in this patient. CONCLUSION We report the first identified case of MSMD among Egyptian patients, including in particular a new IFNGR1 mutation underlying IFN-γR1 deficiency. The eight remaining patients need to be explored further. These findings have implications regarding the compulsory Bacillus.
Collapse
|
42
|
Bustamante J, Picard C, Boisson-Dupuis S, Abel L, Casanova JL. Genetic lessons learned from X-linked Mendelian susceptibility to mycobacterial diseases. Ann N Y Acad Sci 2012; 1246:92-101. [PMID: 22236433 DOI: 10.1111/j.1749-6632.2011.06273.x] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Mendelian susceptibility to mycobacterial disease (MSMD) is a rare syndrome conferring predisposition to clinical disease caused by weakly virulent mycobacteria, such as Mycobacterium bovis Bacille Calmette Guérin (BCG) vaccines and nontuberculous, environmental mycobacteria (EM). Since 1996, MSMD-causing mutations have been found in six autosomal genes involved in IL-12/23-dependent, IFN-γ-mediated immunity. The aim of this review is to provide the description of the two described forms of X-linked recessive (XR) MSMD. Germline mutations in two genes, NEMO and CYBB, have long been known to cause other human diseases-incontinentia pigmenti (IP) and anhidrotic ectodermal dysplasia with immunodeficiency (EDA-ID) (NEMO/IKKG), and X-linked chronic granulomatous disease (CGD) (CYBB)-but specific mutations in either of these two genes have recently been shown to cause XR-MSMD. NEMO is an essential component of several NF-κB-dependent signaling pathways. The MSMD-causing mutations in NEMO selectively affect the CD40-dependent induction of IL-12 in mononuclear cells. CYBB encodes gp91(phox) , which is an essential component of the NADPH oxidase in phagocytes. The MSMD-causing mutation in CYBB selectively affects the respiratory burst in macrophages. Mutations in NEMO and CYBB may therefore cause MSMD by selectively exerting their deleterious impact on a single signaling pathway (CD40-IL-12, NEMO) or a single cell type (macrophages, CYBB). These experiments of Nature illustrate how specific germline mutations in pleiotropic genes can dissociate signaling pathways or cell lineages, thereby resulting in surprisingly narrow clinical phenotypes.
Collapse
Affiliation(s)
- Jacinta Bustamante
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale, Paris, France.
| | | | | | | | | |
Collapse
|
43
|
|
44
|
|
45
|
Infectious diseases in patients with IRAK-4, MyD88, NEMO, or IκBα deficiency. Clin Microbiol Rev 2011; 24:490-7. [PMID: 21734245 DOI: 10.1128/cmr.00001-11] [Citation(s) in RCA: 259] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Autosomal recessive IRAK-4 and MyD88 deficiencies predispose affected patients to recurrent invasive pyogenic bacterial infection. Both defects result in the selective impairment of cellular responses to Toll-like receptors (TLRs) other than TLR3 and of cellular responses to most interleukin-1 receptors (IL-1Rs), including IL-1R, IL-18R, and IL-33R. Hypomorphic mutations in the X-linked NEMO gene and hypermorphic mutations in the autosomal IKBA gene cause X-linked recessive and autosomal dominant anhidrotic ectodermal dysplasia with immunodeficiency (EDA-ID) syndromes. Both of these defects impair NF-κB-mediated cellular responses to multiple receptors, including TLRs, IL-1Rs, and tumor necrosis factor receptors (TNF-Rs). They therefore confer a much broader predisposition to infections than that for IRAK-4 and MyD88 deficiencies. These disorders were initially thought to be rare but have now been diagnosed in over 170 patients worldwide. We review here the infectious diseases affecting patients with inborn errors of NF-κB-dependent TLR and IL-1R immunity.
Collapse
|
46
|
Collin M, Bigley V, Haniffa M, Hambleton S. Human dendritic cell deficiency: the missing ID? Nat Rev Immunol 2011; 11:575-83. [PMID: 21852794 DOI: 10.1038/nri3046] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Animal models and human in vitro systems indicate that dendritic cells (DCs) have a crucial role in priming naive T cells, but just how important are they in the intact human? Recent descriptions of human DC deficiency have begun to shed light on this question and to illuminate other puzzles of human DC biology, including their haematopoietic origin, developmental regulation and homeostatic equilibrium with other leukocytes. In this Review, we explore the recently described DC deficiency syndromes, discussing what these have taught us with regard to DC function in humans and the important issues that remain unsolved.
Collapse
Affiliation(s)
- Matthew Collin
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
| | | | | | | |
Collapse
|
47
|
Songhet P, Barthel M, Stecher B, Müller AJ, Kremer M, Hansson GC, Hardt WD. Stromal IFN-γR-signaling modulates goblet cell function during Salmonella Typhimurium infection. PLoS One 2011; 6:e22459. [PMID: 21829463 PMCID: PMC3145644 DOI: 10.1371/journal.pone.0022459] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Accepted: 06/28/2011] [Indexed: 11/23/2022] Open
Abstract
Enteropathogenic bacteria are a frequent cause of diarrhea worldwide. The mucosal defenses against infection are not completely understood. We have used the streptomycin mouse model for Salmonella Typhimurium diarrhea to analyze the role of interferon gamma receptor (IFN-γR)-signaling in mucosal defense. IFN-γ is known to contribute to acute S. Typhimurium diarrhea. We have compared the acute mucosal inflammation in IFN-γR(-/-) mice and wild type animals. IFN-γR(-/-) mice harbored increased pathogen loads in the mucosal epithelium and the lamina propria. Surprisingly, the epithelium of the IFN-γR(-/-) mice did not show the dramatic "loss" of mucus-filled goblet cell vacuoles, a hallmark of the wild type mucosal infection. Using bone marrow chimeric mice we established that IFN-γR-signaling in stromal cells (e.g. goblet cells, enterocytes) controlled mucus excretion/vacuole loss by goblet cells. In contrast, IFN-γR-signaling in bone marrow-derived cells (e.g. macrophages, DCs, PMNs) was required for restricting pathogen growth in the gut tissue. Thus IFN-γR-signaling influences different mucosal responses to infection, including not only pathogen restriction in the lamina propria, but, as shown here, also goblet cell function.
Collapse
Affiliation(s)
- Pascal Songhet
- Institute of Microbiology (D-BIOL), Eidgenössische Technische Hochschule Zürich, Zürich, Switzerland
| | - Manja Barthel
- Institute of Microbiology (D-BIOL), Eidgenössische Technische Hochschule Zürich, Zürich, Switzerland
| | - Bärbel Stecher
- Institute of Microbiology (D-BIOL), Eidgenössische Technische Hochschule Zürich, Zürich, Switzerland
| | - Andreas J. Müller
- Institute of Microbiology (D-BIOL), Eidgenössische Technische Hochschule Zürich, Zürich, Switzerland
| | - Marcus Kremer
- Institut für Allgemeine Pathologie und Pathologische Anatomie, Technische Universität München, Munich, Germany
| | - Gunnar C. Hansson
- Department of Medical Biochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Wolf-Dietrich Hardt
- Institute of Microbiology (D-BIOL), Eidgenössische Technische Hochschule Zürich, Zürich, Switzerland
| |
Collapse
|
48
|
Abstract
Non-typhoidal Salmonella (NTS) serotypes cause a localized gastroenteritis in immunocompetent individuals. In contrast, primary immunodeficiencies that impair interleukin-23 (IL-23)-dependent pathways are associated in humans with disseminated NTS bloodstream infections (bacteraemia). The recent use of animal models has helped to define the role the IL-23 axis plays during NTS gastroenteritis, but additional work is needed to elucidate how this host defence pathway prevents NTS bacteraemia.
Collapse
Affiliation(s)
- Ivan Godinez
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA, USA
| | | | | | | |
Collapse
|
49
|
Innate Immunity and Host Defense against Microbial Infection. Mol Microbiol 2011. [DOI: 10.1128/9781555816834.ch54] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
50
|
Casanova JL, Abel L, Quintana-Murci L. Human TLRs and IL-1Rs in host defense: natural insights from evolutionary, epidemiological, and clinical genetics. Annu Rev Immunol 2011; 29:447-91. [PMID: 21219179 DOI: 10.1146/annurev-immunol-030409-101335] [Citation(s) in RCA: 246] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Toll-like receptors (TLRs) and interleukin-1 receptors (IL-1Rs) have TIR intracellular domains that engage two main signaling pathways, via the TIR-containing adaptors MyD88 (which is not used by TLR3) and TRIF (which is used only by TLR3 and TLR4). Extensive studies in inbred mice in various experimental settings have attributed key roles in immunity to TLR- and IL-1R-mediated responses, but what contribution do human TLRs and IL-1Rs actually make to host defense in the natural setting? Evolutionary genetic studies have shown that human intracellular TLRs have evolved under stronger purifying selection than surface-expressed TLRs, for which the frequency of missense and nonsense alleles is high in the general population. Epidemiological genetic studies have yet to provide convincing evidence of a major contribution of common variants of human TLRs, IL-1Rs, or their adaptors to host defense. Clinical genetic studies have revealed that rare mutations affecting the TLR3-TRIF pathway underlie herpes simplex virus encephalitis, whereas mutations in the TIR-MyD88 pathway underlie pyogenic bacterial diseases in childhood. A careful reconsideration of the contributions of TLRs and IL-1Rs to host defense in natura is required.
Collapse
Affiliation(s)
- Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, NY 10021, USA.
| | | | | |
Collapse
|