1
|
鲁 玲, 杨 小, 张 华, 梁 媛, 石 秀, 周 鑫. [Recombinant Schistosoma japonicum cystatin alleviates acute liver injury in mice by inhibiting endoplasmic reticulum stress, inflammation and hepatocyte apoptosis]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:1126-1134. [PMID: 38977342 PMCID: PMC11237295 DOI: 10.12122/j.issn.1673-4254.2024.06.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Indexed: 07/10/2024]
Abstract
OBJECTIVE To investigate the protective effect of recombinant Schistosoma japonicum cystatin (rSj-Cys) against acute liver injury induced by lipopolysaccharide (LPS) and D-GalN in mice. METHODS Adult male C57BL/6J mice with or without LPS/D-GaIN-induced acute liver injury were given intraperitoneal injections of rSj-Cys or PBS 30 min after modeling (n=18), and serum and liver tissues samples were collected from 8 mice in each group 6 h after modeling. The survival of the remaining 10 mice in each group within 24 h was observed. Serum levels of ALT, AST, TNF-α and IL-6 of the mice were measured, and liver pathologies was observed with HE staining. The hepatic expressions of macrophage marker CD68, Bax, Bcl-2 and endoplasmic reticulum stress (ERS)-related proteins were detected using immunohistochemistry or immunoblotting, and TUNEL staining was used to detect hepatocyte apoptosis. RESULTS The survival rates of PBS- and rSj-Cys-treated mouse models of acute liver injury were 30% and 80% at 12 h and were 10% and 60% at 24 h after modeling, respectively; no death occurred in the two control groups within 24 h. The mouse models showed significantly increased serum levels of AST, ALT, IL-6 and TNF-α and serious liver pathologies with increased hepatic expressions of CD68 and Bax, lowered expression of Bcl-2, increased hepatocyte apoptosis, and up-regulated expressions of ERS-related signaling pathway proteins GRP78, CHOP and NF-κB p-p65. Treatment of the mouse models significantly lowered the levels of AST, ALT, IL-6 and TNF-α, alleviated liver pathologies, reduced hepatic expressions of CD68, Bax, GRP78, CHOP and NF-κB p-p65, and enhanced the expression of Bcl-2. In the normal control mice, rSj-Cys injection did not produce any significant changes in these parameters compared with PBS. CONCLUSION rSj-Cys alleviates LPS/D-GalN-induced acute liver injury in mice by suppressing ERS, attenuating inflammation and inhibiting hepatocyte apoptosis.
Collapse
|
2
|
Sargsian S, Mondragón-Palomino O, Lejeune A, Ercelen D, Jin WB, Varghese A, Lim YAL, Guo CJ, Loke P, Cadwell K. Functional characterization of helminth-associated Clostridiales reveals covariates of Treg differentiation. MICROBIOME 2024; 12:86. [PMID: 38730492 PMCID: PMC11084060 DOI: 10.1186/s40168-024-01793-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 03/10/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND Parasitic helminths influence the composition of the gut microbiome. However, the microbiomes of individuals living in helminth-endemic regions are understudied. The Orang Asli, an indigenous population in Malaysia with high burdens of the helminth Trichuris trichiura, display microbiotas enriched in Clostridiales, an order of spore-forming obligate anaerobes with immunogenic properties. We previously isolated novel Clostridiales that were enriched in these individuals and found that a subset promoted the Trichuris life cycle. In this study, we aimed to further characterize the functional properties of these bacteria. RESULTS Clostridiales isolates were profiled for their ability to perform 57 enzymatic reactions and produce short-chain fatty acids (SCFAs) and hydrogen sulfide, revealing that these bacteria were capable of a range of activities associated with metabolism and host response. Consistent with this finding, monocolonization of mice with individual isolates identified bacteria that were potent inducers of regulatory T-cell (Treg) differentiation in the colon. Comparisons between variables revealed by these studies identified enzymatic properties correlated with Treg induction and Trichuris egg hatching. CONCLUSION We identified Clostridiales species that are sufficient to induce high levels of Tregs. We also identified a set of metabolic activities linked with Treg differentiation and Trichuris egg hatching mediated by these newly isolated bacteria. Altogether, this study provides functional insights into the microbiotas of individuals residing in a helminth-endemic region. Video Abstract.
Collapse
Affiliation(s)
- Shushan Sargsian
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Octavio Mondragón-Palomino
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Alannah Lejeune
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Defne Ercelen
- Department of Medicine, Division of Gastroenterology and Hepatology, New York University Langone Health, New York, NY, 10016, USA
| | - Wen-Bing Jin
- Weill Cornell Medicine, Jill Roberts Institute for Research in Inflammatory Bowel Disease, Cornell University, New York, NY, 10021, USA
| | - Alan Varghese
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Yvonne A L Lim
- Department of Parasitology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, 50603, Malaysia
| | - Chun-Jun Guo
- Weill Cornell Medicine, Jill Roberts Institute for Research in Inflammatory Bowel Disease, Cornell University, New York, NY, 10021, USA
| | - P'ng Loke
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Ken Cadwell
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
| |
Collapse
|
3
|
Mignini I, Piccirilli G, Termite F, Paratore M, Esposto G, Laterza L, Scaldaferri F, Ainora ME, Gasbarrini A, Zocco MA. Extracellular Vesicles: Novel Potential Therapeutic Agents in Inflammatory Bowel Diseases. Cells 2023; 13:90. [PMID: 38201294 PMCID: PMC10778449 DOI: 10.3390/cells13010090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 12/24/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
Patients affected by inflammatory bowel diseases (IBD) can nowadays benefit from a growing number of pharmacological options. However, in moderate-to-severe cases, the therapeutic response is still far from optimal, and treatment changes and optimizations are often required. Thus, researchers in this field are strongly engaged in studies aiming to identify new potential therapeutic targets. Extracellular vesicles (EVs) are tiny subcellular bodies with a phospholipid bilayer envelope containing bioactive molecules, which are released from different cells and are involved in intercellular communication. Recent pre-clinical data show their emerging role in the pathogenesis and treatment of IBD. In our review, we summarize current evidence about the function of EVs as active therapeutic agents in ulcerative colitis and Crohn's disease, analyzing the properties of EVs derived from different cellular sources and the mechanisms through which they may improve intestinal inflammation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Maria Assunta Zocco
- CEMAD Digestive Diseases Center, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy; (I.M.); (G.P.); (F.T.); (M.P.); (G.E.); (L.L.); (F.S.); (M.E.A.); (A.G.)
| |
Collapse
|
4
|
Wang S, Jiang D, Huang F, Qian Y, Qi M, Li H, Wang X, Wang Z, Wang K, Wang Y, Du P, Zhan B, Zhou R, Chu L, Yang X. Therapeutic effect of Echinococcus granulosus cyst fluid on bacterial sepsis in mice. Parasit Vectors 2023; 16:450. [PMID: 38066526 PMCID: PMC10709918 DOI: 10.1186/s13071-023-06021-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 10/18/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND The primary pathophysiological process of sepsis is to stimulate a massive release of inflammatory mediators to trigger systemic inflammatory response syndrome (SIRS), the major cause of multi-organ dysfunction and death. Like other helminths, Echinococcus granulosus induces host immunomodulation. We sought to determine whether E. granulosus cyst fluid (EgCF) displays a therapeutic effect on sepsis-induced inflammation and tissue damage in a mouse model. METHODS The anti-inflammatory effects of EgCF were determined by in vitro culture with bone marrow-derived macrophages (BMDMs) and in vivo treatment of BALB/C mice with cecal ligation and puncture (CLP)-induced sepsis. The macrophage phenotypes were determined by flow cytometry, and the levels of cytokines in cell supernatants or in sera of mice were measured (ELISA). The therapeutic effect of EgCF on sepsis was evaluated by observing the survival rates of mice for 72 h after CLP, and the pathological injury to the liver, kidney, and lung was measured under a microscope. The expression of TLR-2/MyD88 in tissues was measured by western blot to determine whether TLR-2/MyD88 is involved in the sepsis-induced inflammatory signaling pathway. RESULTS In vitro culture with BMDMs showed that EgCF promoted macrophage polarization to M2 type and inhibited lipopolysaccharide (LPS)-induced M1 macrophages. EgCF treatment provided significant therapeutic effects on CLP-induced sepsis in mice, with increased survival rates and alleviation of tissue injury. The EgCF conferred therapeutic efficacy was associated with upregulated anti-inflammatory cytokines (IL-10 and TGF-β) and reduced pro-inflammatory cytokines (TNF-α and INF-γ). Treatment with EgCF induced Arg-1-expressed M2, and inhibited iNOS-expressed M1 macrophages. The expression of TLR-2 and MyD88 in EgCF-treated mice was reduced. CONCLUSIONS The results demonstrated that EgCF confers a therapeutic effect on sepsis by inhibiting the production of pro-inflammatory cytokines and inducing regulatory cytokines. The anti-inflammatory effect of EgCF is carried out possibly through inducing macrophage polarization from pro-inflammatory M1 to regulatory M2 phenotype to reduce excessive inflammation of sepsis and subsequent multi-organ damage. The role of EgCF in regulating macrophage polarization may be achieved by inhibiting the TLR2/MyD88 signaling pathway.
Collapse
Affiliation(s)
- Shuying Wang
- First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
- Department of Pediatrics, Anqing First People's Hospital of Anhui Medical University, Anqing, 246000, China
| | - Donghui Jiang
- Department of Critical Care Medicine, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Feifei Huang
- First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
| | - Yayun Qian
- First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
| | - Meitao Qi
- Department of Pediatrics, Anqing First People's Hospital of Anhui Medical University, Anqing, 246000, China
| | - Huihui Li
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
| | - Xiaoli Wang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
| | - Zhi Wang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
| | - Kaigui Wang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
| | - Yin Wang
- Department of Critical Care Medicine, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Pengfei Du
- Department of Critical Care Medicine, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Bin Zhan
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Rui Zhou
- First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
| | - Liang Chu
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China.
- Second Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China.
| | - Xiaodi Yang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China.
| |
Collapse
|
5
|
Li Z, Wang X, Zhang W, Yang W, Xu B, Hu W. Excretory/Secretory Products from Schistosoma japonicum Eggs Alleviate Ovalbumin-Induced Allergic Airway Inflammation. PLoS Negl Trop Dis 2023; 17:e0011625. [PMID: 37788409 PMCID: PMC10547495 DOI: 10.1371/journal.pntd.0011625] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 08/29/2023] [Indexed: 10/05/2023] Open
Abstract
INTRODUCTION Excretory/secretory products (ESPs) derived from helminths have been reported to effectively control allergic inflammation, which have better therapeutic prospects than live parasite infections. However, it remains unknown whether ESPs from schistosome eggs can protect against allergies, despite reports alleging that schistosome infection could alleviate disordered allergic inflammation. METHOD In the present study, we investigated the protective effects of ESPs from Schistosoma japonicum eggs (ESP-SJE) on asthmatic inflammation. Firstly, we successfully established an allergic airway inflammation model in mice by alum-adjuvanted ovalbumin (OVA) sensitization and challenge. ESP-SJE were administered intraperitoneally on days -1 and 13 (before sensitization), on day 20 (before challenge), and on days 21-24 (challenge phase). RESULTS The results showed that ESP-SJE treatment significantly reduced the infiltration of inflammatory cells, especially eosinophils into the lung tissue, inhibited the production of the total and OVA-specific IgE during OVA-sensitized and -challenged phases, respectively, and suppressed the secretion of Th2-type inflammatory cytokines (IL-4). Additionally, ESP-SJE treatment significantly upregulated the regulatory T cells (Tregs) in the lung tissue during OVA challenge. Furthermore, using liquid chromatography-mass spectrometry analysis and Treg induction experiments in vitro, we might identify nine potential therapeutic proteins against allergic inflammation in ESP-SJE. The targets of these candidate proteins included glutathione S-transferase, egg protein CP422 precursor, tubulin alpha-2/alpha-4 chain, actin-2, T-complex protein 1 subunit beta, histone H₄, whey acidic protein core region, and molecular chaperone HtpG. CONCLUSION Taken together, the results discussed herein demonstrated that ESP-SJE could significantly alleviate OVA-induced asthmatic inflammation in a murine model, which might be mediated by the upregulation of Treg in lung tissues that may be induced by the potential modulatory proteins. Therefore, potential proteins in ESP-SJE might be the best candidates to be tested for therapeutic application of asthma, thus pointing out to a possible new therapy for allergic airway inflammation.
Collapse
Affiliation(s)
- Zhidan Li
- Department of Immunology, Binzhou Medical University, Yantai, Shandong, P. R. China
- National Institute of Parasitic Diseases, Chinese Centre for Disease Control and Prevention, WHO Collaborating Centre for Tropical Diseases, National Centre for International Research on Tropical Diseases, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health, Shanghai, China
| | - Xiaoling Wang
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, Human Phenome Institute, Ministry of Education Key Laboratory for Biodiversity Science and Ecological Engineering, Department of Microbiology and Microbial Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Wei Zhang
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, Human Phenome Institute, Ministry of Education Key Laboratory for Biodiversity Science and Ecological Engineering, Department of Microbiology and Microbial Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Wenbin Yang
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, Human Phenome Institute, Ministry of Education Key Laboratory for Biodiversity Science and Ecological Engineering, Department of Microbiology and Microbial Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Bin Xu
- National Institute of Parasitic Diseases, Chinese Centre for Disease Control and Prevention, WHO Collaborating Centre for Tropical Diseases, National Centre for International Research on Tropical Diseases, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health, Shanghai, China
| | - Wei Hu
- National Institute of Parasitic Diseases, Chinese Centre for Disease Control and Prevention, WHO Collaborating Centre for Tropical Diseases, National Centre for International Research on Tropical Diseases, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health, Shanghai, China
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, Human Phenome Institute, Ministry of Education Key Laboratory for Biodiversity Science and Ecological Engineering, Department of Microbiology and Microbial Engineering, School of Life Sciences, Fudan University, Shanghai, China
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
- College of Life Sciences, Inner Mongolia University, Hohhot, China
| |
Collapse
|
6
|
Sargsian S, Lejeune A, Ercelen D, Jin WB, Varghese A, Loke P, Lim YAL, Guo CJ, Cadwell K. Functional characterization of helminth-associated Clostridiales reveals covariates of Treg differentiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.05.543751. [PMID: 37333296 PMCID: PMC10274677 DOI: 10.1101/2023.06.05.543751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Parasitic helminths influence the composition of the gut microbiome. However, the microbiomes of individuals living in helminth-endemic regions are understudied. The Orang Asli, an indigenous population in Malaysia with high burdens of the helminth Trichuris trichiura, displayed microbiotas enriched in Clostridiales, an order of spore-forming obligate anaerobes previously shown to have immunogenic properties. We previously isolated novel Clostridiales that were enriched in these individuals and found that a subset promoted the Trichuris life cycle. Here, we further characterized the functional properties of these bacteria. Enzymatic and metabolomic profiling revealed a range of activities associated with metabolism and host response. Consistent with this finding, monocolonization of mice with individual isolates identified bacteria that were potent inducers of regulatory T cell (Treg) differentiation in the colon. Comparisons between variables revealed by these studies identified enzymatic properties correlated with Treg induction and Trichuris egg hatching. These results provide functional insights into the microbiotas of an understudied population.
Collapse
Affiliation(s)
- Shushan Sargsian
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Alannah Lejeune
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Defne Ercelen
- Division of Gastroenterology and Hepatology, Department of Medicine, New York University Langone Health, New York, NY 10016, USA
| | - Wen-Bing Jin
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Alan Varghese
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - P’ng Loke
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yvonne A. L. Lim
- Department of Parasitology, Faculty of Medicine, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Chun-Jun Guo
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Ken Cadwell
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
- Lead Contact
| |
Collapse
|
7
|
Suresh KP, Indrabalan UB, Shreevatsa B, Dharmashekar C, Singh P, Patil SS, Syed A, Elgorban AM, Eswaramoorthy R, Amachawadi RG, Shivamallu C, Kollur SP. Evaluation of codon usage patterns and molecular evolution dynamics in Japanese encephalitis virus: An integrated bioinformatics approach. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2023; 109:105410. [PMID: 36791944 DOI: 10.1016/j.meegid.2023.105410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 01/04/2023] [Accepted: 02/07/2023] [Indexed: 02/16/2023]
Abstract
In the recent survey, Japanese encephalitis (JE) is one of the most common mosquito-borne diseases, accounting for ∼30% of fatalities. The outbreaks of the JE virus (JEV) suggests that exhaustive study is essential for the prevention and management of the disease. The disease mainly spreads in humans and pigs by the vector: mosquito; as this is a major concern, this study had employed various bioinformatics tools to investigate the codon usage bias, evolutionary inference and selection pressure analysis of the Japanese encephalitis virus disease. The results indicated that the JE virus was biased and natural selection was the main factor shaping the codon usage that was determined and confirmed with the Nc, neutrality, PR2 plots and correlation analysis. The evolutionary analysis revealed that the virus had a substitution rate of 1.54 × 10-4 substitution/site/year and the tMRCA was found to be in 1723. The transmission of the virus in the map found transmissions mostly from China and transmitted across Asia and Africa. The selection pressure analysis employed three methods which had 969th codon site as diversifying site and had many purifying sites that shows the virus had evolved rapidly. The inferences from this study would aid people to employ this methodology on various diseases and also perform insilico studies in the field of vaccinology and immunoinformatics.
Collapse
Affiliation(s)
| | - Uma Bharathi Indrabalan
- ICAR-National Institute of Veterinary Epidemiology and Disease Informatics (NIVEDI), Yelahanka, Bengaluru, Karnataka 560063, India
| | - Bhargav Shreevatsa
- Department of Biotechnology and Bioinformatics, School of Life Sciences, JSS Academy of Higher Education & Research, Myuru 570015, Karnataka, India.
| | - Chandan Dharmashekar
- Department of Biotechnology and Bioinformatics, School of Life Sciences, JSS Academy of Higher Education & Research, Myuru 570015, Karnataka, India.
| | - Pranav Singh
- Department of Internal Medicine, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Sharanagouda S Patil
- ICAR-National Institute of Veterinary Epidemiology and Disease Informatics (NIVEDI), Yelahanka, Bengaluru, Karnataka 560063, India.
| | - Asad Syed
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. 2455, Riyadh 11451, Saudi Arabia.
| | - Abdallah M Elgorban
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. 2455, Riyadh 11451, Saudi Arabia.
| | - Rajalakshmanan Eswaramoorthy
- Department of Prosthodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai 600 077, Tamilnadu, India.
| | - Raghavendra G Amachawadi
- Department of Clinical Sciences, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506-5606, USA.
| | - Chandan Shivamallu
- Department of Biotechnology and Bioinformatics, School of Life Sciences, JSS Academy of Higher Education & Research, Myuru 570015, Karnataka, India.
| | - Shiva Prasad Kollur
- School of Physical Sciences, Amrita Vishwa Vidhyapeetham, Mysuru Campus, Mysuru 570 026, Karnataka, India.
| |
Collapse
|
8
|
Rovira-Diaz E, El-Naccache DW, Reyes J, Zhao Y, Nasuhidehnavi A, Chen F, Gause WC, Yap GS. The Impact of Helminth Coinfection on Innate and Adaptive Immune Resistance and Disease Tolerance during Toxoplasmosis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:2160-2171. [PMID: 36426972 PMCID: PMC10065986 DOI: 10.4049/jimmunol.2200504] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 09/23/2022] [Indexed: 01/04/2023]
Abstract
More than 2 billion people worldwide are infected with helminths. Thus, it is possible for individuals to experience concomitant infection with helminth and intracellular microbes. Although the helminth-induced type 2 response can suppress type 1 proinflammatory responses required for the immunity against intracellular pathogens in the context of a coinfection, conflicting evidence suggest that helminth infection can enhance antimicrobial immunity. Using a coinfection model with the intestinal helminth Heligmosomoides polygyrus followed by infection with Toxoplasma gondii in Mus Musculus, we showed that the complex and dynamic effect of helminth infection is highly suppressive during the innate phase (days 0-3) of T. gondii infection and less stringent during the acute phase (d10). Helminth coinfection had a strong suppressive effect on the neutrophil, monocytic, and early IFN-γ/IL-12 responses. The IFN-γ response was later restored by compensatory production from T cells despite decreased effector differentiation of T. gondii-specific CD8 T cells. In accordance with the attenuated IFN-γ response, parasite loads were elevated during the acute phase (d10) of T. gondii infection but were transiently controlled by the compensatory T cell response. Unexpectedly, 40% of helminth-coinfected mice exhibited a sustained weight loss phenotype during the postacute phase (d14-18) that was not associated with T. gondii outgrowth, indicating that coinfection led to decreased disease tolerance during T. gondii infection. Our work uncovers the dynamic nature of the helminth immunomodulatory effects on concomitant infections or immune responses and unveils a loss of disease tolerance phenotype triggered by coinfection with intestinal helminth.
Collapse
Affiliation(s)
- Eliezer Rovira-Diaz
- Department of Medicine and Center for Immunity and Inflammation, New Jersey Medical School, Rutgers University, Newark, NJ 07101
| | - Darine W. El-Naccache
- Department of Medicine and Center for Immunity and Inflammation, New Jersey Medical School, Rutgers University, Newark, NJ 07101
| | - Jojo Reyes
- Department of Medicine and Center for Immunity and Inflammation, New Jersey Medical School, Rutgers University, Newark, NJ 07101
| | - Yanlin Zhao
- Department of Medicine and Center for Immunity and Inflammation, New Jersey Medical School, Rutgers University, Newark, NJ 07101
| | - Azadeh Nasuhidehnavi
- Department of Medicine and Center for Immunity and Inflammation, New Jersey Medical School, Rutgers University, Newark, NJ 07101
| | - Fei Chen
- Department of Medicine and Center for Immunity and Inflammation, New Jersey Medical School, Rutgers University, Newark, NJ 07101
| | - William C. Gause
- Department of Medicine and Center for Immunity and Inflammation, New Jersey Medical School, Rutgers University, Newark, NJ 07101
| | - George S. Yap
- Department of Medicine and Center for Immunity and Inflammation, New Jersey Medical School, Rutgers University, Newark, NJ 07101
| |
Collapse
|
9
|
Sargsian S, Chen Z, Lee SC, Robertson A, Thur RS, Sproch J, Devlin JC, Tee MZ, Er YX, Copin R, Heguy A, Pironti A, Torres VJ, Ruggles KV, Lim YA, Bethony J, Loke P, Cadwell K. Clostridia isolated from helminth-colonized humans promote the life cycle of Trichuris species. Cell Rep 2022; 41:111725. [PMID: 36450245 PMCID: PMC9790084 DOI: 10.1016/j.celrep.2022.111725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 08/31/2022] [Accepted: 11/03/2022] [Indexed: 12/03/2022] Open
Abstract
Soil-transmitted intestinal worms known as helminths colonize over 1.5 billion people worldwide. Although helminth colonization has been associated with altered composition of the gut microbiota, such as increases in Clostridia, individual species have not been isolated and characterized. Here, we isolate and sequence the genome of 13 Clostridia from the Orang Asli, an indigenous population in Malaysia with a high prevalence of helminth infections. Metagenomic analysis of 650 fecal samples from urban and rural Malaysians confirm the prevalence of species corresponding to these isolates and reveal a specific association between Peptostreptococcaceae family members and helminth colonization. Remarkably, Peptostreptococcaceae isolated from the Orang Asli display superior capacity to promote the life cycle of whipworm species, including hatching of eggs from Trichuris muris and Trichuris trichiura. These findings support a model in which helminths select for gut colonization of microbes that support their life cycle.
Collapse
Affiliation(s)
- Shushan Sargsian
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University Grossman School of Medicine, New York, NY 10016, USA,Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Ze Chen
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Soo Ching Lee
- Type 2 Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Amicha Robertson
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University Grossman School of Medicine, New York, NY 10016, USA,Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Rafaela Saes Thur
- Department of Microbiology, Immunology and Tropical Medicine, George Washington University, Washington, DC 20052, USA
| | - Julia Sproch
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Joseph C. Devlin
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Mian Zi Tee
- Department of Biomedical Science, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Yi Xian Er
- Department of Parasitology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Richard Copin
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Adriana Heguy
- Genome Technology Center, Office of Science and Research, New York University Langone Health, New York, NY 10016, USA,Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Alejandro Pironti
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA,Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, NY 10016, USA,Microbial Computational Genomic Core Lab, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Victor J. Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA,Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Kelly V. Ruggles
- Institute for System Genetics, New York University Langone Health, New York, NY 10016, USA,Division of Precision Medicine, Department of Medicine, New York University Langone Health, New York, NY 10016, USA
| | - Yvonne A.L. Lim
- Department of Parasitology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Jeffrey Bethony
- Department of Microbiology, Immunology and Tropical Medicine, George Washington University, Washington, DC 20052, USA
| | - P’ng Loke
- Type 2 Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA,Correspondence: (P.L.), (K.C.)
| | - Ken Cadwell
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University Grossman School of Medicine, New York, NY 10016, USA,Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA,Division of Gastroenterology and Hepatology, Department of Medicine, New York University Langone Health, New York, NY 10016, USA,Lead contact,Correspondence: (P.L.), (K.C.)
| |
Collapse
|
10
|
Razafiarimanga ZN, Yao YBK, Rajerison M, Randriamampianina LJ, Rahelinirina S, Rakotoarison R, Alexandra B, Elisoa H, Pascal H, Ronan J. Risk factors for intestinal parasite portage in an informal suburb on the West coast of Madagascar. Parasite Epidemiol Control 2022; 19:e00267. [PMID: 36065443 PMCID: PMC9440058 DOI: 10.1016/j.parepi.2022.e00267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 06/21/2022] [Accepted: 08/15/2022] [Indexed: 10/31/2022] Open
Abstract
The deprived area of the Metzinger Valley in the city of Mahajanga has many healthcare concerns due to repeated flooding during the rainy season. Improving this health situation requires a better knowledge of the pathogens present in this area and of the risk factors favoring their propagation. The aim of this study was to analyze the relationship between the household socioeconomic status and the presence of parasites in the faeces of children between 1 and 10 years of age in order to determine the risk factors for intestinal parasitosis. The study included 746 children, of whom 30% were infected with parasites. Entamoeba coli, a good indicator of environmental fecal contamination, was the most prevalent parasite with an observation frequency of 16.7% followed by Giardia lamblia with a prevalence of 10%. For helminths, Trichuris and Ascaris were the most prevalent respectively 5.4% and 1.8%. A large heterogeneity in the prevalence of parasites was observed from one neighborhood to another. However, multivariate analysis showed that these differences were not related to environmental factors or household structure, but rather to the economic level of the family, the education level of the mother as well as the age of the child. For example, the prevalence of Giardia decreased from 23.5% to 8% for children of mothers with little education to those with higher education, respectively. For E. coli, the prevalence is higher among poor households and school-aged children. In the frame of IRCOD project, mothers are being sensitized to hygiene and risk factors for transmission by intestinal parasites and the present study proposes a multidimensional approach as an assessment tool.
Collapse
Affiliation(s)
| | - Yves Boris Kouakou Yao
- Department of Parasitology Mycology, Institut Pasteur de Côte d'Ivoire, Abidjan, Cote d'Ivoire
| | | | | | | | | | - Bastaraud Alexandra
- Food and water Unit Institut Pasteur de Madagascar, Antananarivo, Madagascar
| | - Hariniaina Elisoa
- Clinical Biology Laboratory Institut Pasteur de Madagascar, Antananarivo, Madagascar
| | | | - Jambou Ronan
- Department of Parasitology Mycology, Institut Pasteur de Côte d'Ivoire, Abidjan, Cote d'Ivoire
- Global Health Department, Institut Pasteur, Paris, France
| |
Collapse
|
11
|
Huang H, Hu D, Chen Z, Xu J, Xu R, Gong Y, Fang Z, Wang T, Chen W. Immunotherapy for type 1 diabetes mellitus by adjuvant-free Schistosoma japonicum-egg tip-loaded asymmetric microneedle patch (STAMP). J Nanobiotechnology 2022; 20:377. [PMID: 35964125 PMCID: PMC9375265 DOI: 10.1186/s12951-022-01581-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 08/01/2022] [Indexed: 11/23/2022] Open
Abstract
Background Type 1 diabetes mellitus (T1DM) is an autoimmune disease mediated by autoreactive T cells and dominated by Th1 response polarization. Insulin replacement therapy faces great challenges to this autoimmune disease, requiring highly frequent daily administration. Intriguingly, the progression of T1DM has proven to be prevented or attenuated by helminth infection or worm antigens for a relatively long term. However, the inevitable problems of low safety and poor compliance arise from infection with live worms or direct injection of antigens. Microneedles would be a promising candidate for local delivery of intact antigens, thus providing an opportunity for the clinical immunotherapy of parasitic products. Methods We developed a Schistosoma japonicum-egg tip-loaded asymmetric microneedle patch (STAMP) system, which serves as a new strategy to combat TIDM. In order to improve retention time and reduce contamination risk, a specific imperfection was introduced on the STAMP (asymmetric structure), which allows the tip to quickly separate from the base layer, improving reaction time and patient’s comfort. After loading Schistosoma japonicum-egg as the immune regulator, the effects of STAMP on blood glucose control and pancreatic pathological progression improvement were evaluated in vivo. Meanwhile, the immunoregulatory mechanism and biosafety of STAMP were confirmed by histopathology, qRT-PCR, ELISA and Flow cytometric analysis. Results Here, the newly developed STAMP was able to significantly reduce blood glucose and attenuate the pancreatic injury in T1DM mice independent of the adjuvants. The isolated Schistosoma japonicum-eggs micron slowly degraded in the skin and continuously released egg antigen for at least 2 weeks, ensuring localization and safety of antigen stimulation. This phenomenon should be attributed to the shift of Th2 immune response to reduce Th1 polarization. Conclusion Our results exhibited that STAMP could significantly regulate the blood glucose level and attenuate pancreatic pathological injury in T1DM mice by balancing the Th1/Th2 immune responses, which is independent of adjuvants. This technology opens a new window for the application of parasite products in clinical immunotherapy. Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01581-9.
Collapse
Affiliation(s)
- Haoming Huang
- National Demonstration Center for Experimental Basic Medical Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Dian Hu
- National Demonstration Center for Experimental Basic Medical Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Zhuo Chen
- National Demonstration Center for Experimental Basic Medical Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Jiarong Xu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Rengui Xu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Yusheng Gong
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Zhengming Fang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Ting Wang
- National Demonstration Center for Experimental Basic Medical Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China. .,Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - Wei Chen
- National Demonstration Center for Experimental Basic Medical Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China. .,Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China. .,Hubei Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
12
|
Yang H, Li H, Chen W, Mei Z, Yuan Y, Wang X, Chu L, Xu Y, Sun Y, Li D, Gao H, Zhan B, Li H, Yang X. Therapeutic Effect of Schistosoma japonicum Cystatin on Atherosclerotic Renal Damage. Front Cell Dev Biol 2021; 9:760980. [PMID: 34901005 PMCID: PMC8656285 DOI: 10.3389/fcell.2021.760980] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 10/11/2021] [Indexed: 01/15/2023] Open
Abstract
Atherosclerosis is a chronic inflammation of the arterial vessel wall driven by lipid metabolism disorders. Although helminthic infection and their derivatives have been identified to attenuate the chronic inflammatory diseases, the immunomodulatory effect of recombinant Schistosoma japonicum cystatin (rSj-Cys) on metabolic diseases and atherosclerosis has not been reported. In this study, we investigated the therapeutic efficacy of rSj-Cys on atherosclerotic renal damage and explored the related immunological mechanism. The results demonstrated that treatment with rSj-Cys significantly reduced body weight gain, hyperlipidemia, and atherosclerosis induced by the high-fat diet in apoE–/– mice. The treatment of rSj-Cys also significantly improved kidney functions through promoting macrophage polarization from M1 to M2, therefore inhibiting M1 macrophage–induced inflammation. The possible mechanism underlying the regulatory effect of rSj-Cys on reducing atherosclerosis and atherosclerotic renal damage is that rSj-Cys stimulates regulatory T cell and M2 macrophage polarization that produce regulatory cytokines, such as interleukin 10 and transforming growth factor β. The therapeutic effect of rSj-Cys on atherosclerotic renal damage is possibly through inhibiting the activation of TLR2/Myd88 signaling pathway. The results in this study provide evidence for the first time that Schistosoma-derived cystatin could be developed as a therapeutic agent to treat lipid metabolism disorder and atherosclerosis that threats million lives around the world.
Collapse
Affiliation(s)
- Huijuan Yang
- Department of Nephrology, First Affiliated Hospital of Bengbu Medical College, Bengbu, China.,Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China
| | - Hongqi Li
- Department of Gerontology, Anhui Provincial Hospital, First Affiliated Hospital of University of Science and Technology of China, Hefei, China
| | - Weidong Chen
- Department of Nephrology, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Zhijie Mei
- Department of Urology, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yuan Yuan
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Basic Medical College of Bengbu Medical College, Bengbu, China
| | - Xiaoli Wang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Basic Medical College of Bengbu Medical College, Bengbu, China
| | - Liang Chu
- Second Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yu Xu
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Basic Medical College of Bengbu Medical College, Bengbu, China
| | - Yan Sun
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Basic Medical College of Bengbu Medical College, Bengbu, China
| | - Dingru Li
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Basic Medical College of Bengbu Medical College, Bengbu, China
| | - Hongyu Gao
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Basic Medical College of Bengbu Medical College, Bengbu, China
| | - Bin Zhan
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Huihui Li
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Basic Medical College of Bengbu Medical College, Bengbu, China
| | - Xiaodi Yang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Basic Medical College of Bengbu Medical College, Bengbu, China
| |
Collapse
|
13
|
Verma U, Ashopa V, Nareda P, Gupta E, Gupta E, Prakash P. " Strongyloides stercoralis infestation in a severely malnourished (SAM) celiac disease child:" A rare case report from Western Rajasthan. Trop Parasitol 2021; 11:60-63. [PMID: 34195065 PMCID: PMC8213110 DOI: 10.4103/tp.tp_86_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 01/05/2021] [Accepted: 01/15/2021] [Indexed: 11/04/2022] Open
Abstract
A case of Strongyloides stercoralis infection in severe protein energy malnutrition child with associated celiac disease (CD) is herein reported. The case was a 4-year-old, severely malnourished female admitted to the tertiary level hospital of Western Rajasthan, due to watery diarrhea, pain abdomen, and vomiting, not responding to treatment. The patient was HIV negative, nondiabetic, had no evidence of tuberculosis, Liver Function Test, Renal Function Test were normal was within normal limits. She had microcytic hypochromic anemia, hypoalbuminemia, and serum Antitissue transglutaminase (anti tTG IgA)was 301.35 U/ML which confirmed the diagnosis of CD. Stool parasitological examination revealed numerous rhabditiform larvae of threadworm "S. stercoralis." After treatment with gluten free diet and Albendazole and Ivermectin, the patient recovered without evidence of S. stercoralis in follow-up stool samples.
Collapse
Affiliation(s)
- Usha Verma
- Dr. S.N. Medical College, Jodhpur, Rajasthan, India
| | | | - Pooja Nareda
- Dr. S.N. Medical College, Jodhpur, Rajasthan, India
| | - Eshank Gupta
- Dr. S.N. Medical College, Jodhpur, Rajasthan, India
| | - Ekta Gupta
- Department of Orthodontics, AIIMS, Jodhpur, Rajasthan, India
| | | |
Collapse
|
14
|
Long SR, Liu RD, Kumar DV, Wang ZQ, Su CW. Immune Protection of a Helminth Protein in the DSS-Induced Colitis Model in Mice. Front Immunol 2021; 12:664998. [PMID: 33995396 PMCID: PMC8117093 DOI: 10.3389/fimmu.2021.664998] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 04/08/2021] [Indexed: 12/23/2022] Open
Abstract
Inflammatory bowel disease (IBD) increases the risk of colorectal cancer, and it has the potential to diminish the quality of life. Recent clinical and experimental evidence demonstrate protective aspects of parasitic helminth infection against IBD. Reports have highlighted the potential use of helminths and their byproducts as potential treatment for IBD. In the current study, we studied the effect of a newborn larvae-specific serine protease from Trichinella spiralis (TsSp) on the host immune and inflammatory responses. A 49-kDa recombinant TsSp (rTsSp) was expressed in Escherichia coli BL21 (DE3) and purified. The cytotoxicity of rTsSp was analyzed. The immune protective effect of rTsSp was studied by using dextran sodium sulfate (DSS)-induced mouse colitis model. The result illustrated that rTsSp has no toxic effects on cells. We further demonstrated that administration of the rTsSp without the additional adjuvant before the induction of DSS-induced colitis reduced the severity of intestinal inflammation and the disease index; it suppressed macrophage infiltration, reduced TNF-α secretion, and induced IL-10 expression. Our findings suggest therapeutic potential of rTsSp on colitis by altering the effect of macrophages. Data also suggest immunotherapy with rTsSp holds promise for use as an additional strategy to positively modulate inflammatory processes involved in IBD.
Collapse
Affiliation(s)
- Shao Rong Long
- Department of Parasitology, Medical College of Zhengzhou University, Zhengzhou, China.,Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Ruo Dan Liu
- Department of Parasitology, Medical College of Zhengzhou University, Zhengzhou, China
| | - Deepak Vijaya Kumar
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Zhong Quan Wang
- Department of Parasitology, Medical College of Zhengzhou University, Zhengzhou, China
| | - Chien-Wen Su
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| |
Collapse
|
15
|
Rosa BA, Snowden C, Martin J, Fischer K, Kupritz J, Beshah E, Supali T, Gankpala L, Fischer PU, Urban JF, Mitreva M. Whipworm-Associated Intestinal Microbiome Members Consistent Across Both Human and Mouse Hosts. Front Cell Infect Microbiol 2021; 11:637570. [PMID: 33777847 PMCID: PMC7991909 DOI: 10.3389/fcimb.2021.637570] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 02/09/2021] [Indexed: 12/14/2022] Open
Abstract
The human whipworm Trichuris trichiura infects 289 million people worldwide, resulting in substantial morbidity. Whipworm infections are difficult to treat due to low cure rates and high reinfection rates. Interactions between whipworm and its host's intestinal microbiome present a potential novel target for infection control or prevention but are very complicated and are identified using inconsistent methodology and sample types across the literature, limiting their potential usefulness. Here, we used a combined 16S rRNA gene OTU analysis approach (QIIME2) for samples from humans and mice infected with whipworm (T. trichiura and T. muris, respectively) to identify for the first time, bacterial taxa that were consistently associated with whipworm infection spanning host species and infection status using four independent comparisons (baseline infected vs uninfected and before vs after deworming for both humans and mice). Using these four comparisons, we identified significant positive associations for seven taxa including Escherichia, which has been identified to induce whipworm egg hatching, and Bacteroides, which has previously been identified as a major component of the whipworm internal microbiome. We additionally identified significant negative associations for five taxa including four members of the order Clostridiales, two from the family Lachnospiraceae, including Blautia which was previously identified as positively associated with whipworm in independent human and mouse studies. Using this approach, bacterial taxa of interest for future association and mechanistic studies were identified, and several were validated by RT-qPCR. We demonstrate the applicability of a mouse animal model for comparison to human whipworm infections with respect to whipworm-induced intestinal microbiome disruption and subsequent restoration following deworming. Overall, the novel cross-species analysis approach utilized here provides a valuable research tool for studies of the interaction between whipworm infection and the host intestinal microbiome.
Collapse
Affiliation(s)
- Bruce A. Rosa
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Caroline Snowden
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - John Martin
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Kerstin Fischer
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Jonah Kupritz
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Ethiopia Beshah
- U.S. Department of Agriculture, Agricultural Research Service, Beltsville Agricultural Research Center, Animal Parasitic Diseases Laboratory, Beltsville, MD, United States
| | - Taniawati Supali
- Department of Parasitology, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Lincoln Gankpala
- Public Health and Medical Research, National Public Health Institute of Liberia, Charlesville, Liberia
| | - Peter U. Fischer
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Joseph F. Urban
- U.S. Department of Agriculture, Agricultural Research Service, Beltsville Agricultural Research Center, Animal Parasitic Diseases Laboratory, Beltsville, MD, United States
| | - Makedonka Mitreva
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, United States
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
16
|
Yordanova IA, Ebner F, Schulz AR, Steinfelder S, Rosche B, Bolze A, Paul F, Mei HE, Hartmann S. The Worm-Specific Immune Response in Multiple Sclerosis Patients Receiving Controlled Trichuris suis Ova Immunotherapy. Life (Basel) 2021; 11:life11020101. [PMID: 33572978 PMCID: PMC7912101 DOI: 10.3390/life11020101] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/19/2021] [Accepted: 01/25/2021] [Indexed: 12/19/2022] Open
Abstract
Considering their potent immunomodulatory properties, therapeutic applications of Trichuris suis ova (TSO) are studied as potential alternative treatment of autoimmune disorders like multiple sclerosis (MS), rheumatoid arthritis (RA), or inflammatory bowel disease (IBD). Clinical phase 1 and 2 studies have demonstrated TSO treatment to be safe and well tolerated in MS patients, however, they reported only modest clinical efficacy. We therefore addressed the cellular and humoral immune responses directed against parasite antigens in individual MS patients receiving controlled TSO treatment (2500 TSO p.o. every 2 weeks for 12 month). Peripheral blood mononuclear cells (PBMC) of MS patients treated with TSO (n = 5) or placebo (n = 6) were analyzed. A continuous increase of serum IgG and IgE antibodies specific for T. suis excretory/secretory antigens was observed up to 12 months post-treatment. This was consistent with mass cytometry analysis identifying an increase of activated HLA-DRhigh plasmablast frequencies in TSO-treated patients. While stable and comparable frequencies of total CD4+ and CD8+ T cells were detected in placebo and TSO-treated patients over time, we observed an increase of activated HLA-DR+CD4+ T cells in TSO-treated patients only. Frequencies of Gata3+ Th2 cells and Th1/Th2 ratios remained stable during TSO treatment, while Foxp3+ Treg frequencies varied greatly between individuals. Using a T. suis antigen-specific T cell expansion assay, we also detected patient-to-patient variation of antigen-specific T cell recall responses and cytokine production. In summary, MS patients receiving TSO treatment established a T. suis-specific T- and B-cell response, however, with varying degrees of T cell responses and cellular functionality across individuals, which might account for the overall miscellaneous clinical efficacy in the studied patients.
Collapse
Affiliation(s)
- Ivet A. Yordanova
- Institute of Immunology, Center for Infection Medicine, Freie Universität Berlin, D-14163 Berlin, Germany; (I.A.Y.); (F.E.)
| | - Friederike Ebner
- Institute of Immunology, Center for Infection Medicine, Freie Universität Berlin, D-14163 Berlin, Germany; (I.A.Y.); (F.E.)
| | - Axel Ronald Schulz
- German Rheumatism Research Center Berlin (DRFZ), a Leibniz Institute, D-10117 Berlin, Germany; (A.R.S.); (H.E.M.)
| | | | - Berit Rosche
- Department of Neurology and Experimental Neurology, Charité—Universitätsmedizin Berlin, D-10117 Berlin, Germany;
- Clinical and Experimental Multiple Sclerosis Research Center, Charité—Universitätsmedizin Berlin, D-10117 Berlin, Germany;
| | - Anna Bolze
- NeuroCure Clinical Research Center, Charité—Universitätsmedizin Berlin, D-10117 Berlin, Germany;
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine, Charité—Universitätsmedizin Berlin, D-10117 Berlin, Germany
| | - Friedemann Paul
- Clinical and Experimental Multiple Sclerosis Research Center, Charité—Universitätsmedizin Berlin, D-10117 Berlin, Germany;
- NeuroCure Clinical Research Center, Charité—Universitätsmedizin Berlin, D-10117 Berlin, Germany;
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine, Charité—Universitätsmedizin Berlin, D-10117 Berlin, Germany
| | - Henrik E. Mei
- German Rheumatism Research Center Berlin (DRFZ), a Leibniz Institute, D-10117 Berlin, Germany; (A.R.S.); (H.E.M.)
| | - Susanne Hartmann
- Institute of Immunology, Center for Infection Medicine, Freie Universität Berlin, D-14163 Berlin, Germany; (I.A.Y.); (F.E.)
- Correspondence:
| |
Collapse
|
17
|
Badri M, Ghaffarifar F, Hassan ZM, Dalimi A, Cortes H. Immunoregulatory Effects of Somatic Extract of Toxocara canis on Airway Inflammations in Murine Model. IRANIAN JOURNAL OF PARASITOLOGY 2020; 15:500-510. [PMID: 33884007 PMCID: PMC8039495 DOI: 10.18502/ijpa.v15i4.4855] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Background: The immunomodulatory role of many parasites is well-documented. The current study designed to assess the immunoregulatory effects of the somatic extract (SE) of Toxocara canis on murine model of airway inflammations. Methods: The experiment was performed in department of parasitology of Tarbiat Mo-dares University, Tehran, Iran from November 2018 to May 2019. Totally 30 female BALB/c mice divided into one control group and two experimental groups (10 mice in each group). The ovalbumin (OVA) group was sensitized with OVA in alum, while the SE group was administered with SE and OVA in alum intraperitoneally. The control group was injected with PBS in alum. Then, SE and OVA groups were intranasally challenged with OVA for three consecutive days and the control group encountered with PBS at the same time. One day after the last challenge, real-time PCR and histopathology survey were conducted on isolated lung tissues. Results: The gene expression of IL-25, IL-33, TNF-α and TLR-4 in SE group was significantly lower than OVA group (P<0.05). The level of IL-10, TGF-β and IFN-γ were considerably higher than the OVA group (P<0.05). The inflammation was reduced in SE group, as the total cell number of bronchoalveolar lavage fluid was less than OVA group. Based on the histopathology findings the inflammation was decreased in SE group compared to the OVA group. Conclusion: Although, an inhibitory effect of SE of T. canis on airway inflammations was detected, there is still a long way ahead regarding the indication of the precise mechanisms.
Collapse
Affiliation(s)
- Milad Badri
- Department of Parasitology and Entomology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Ghaffarifar
- Department of Parasitology and Entomology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Zuhair M Hassan
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Abdolhossein Dalimi
- Department of Parasitology and Entomology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hélder Cortes
- Victor Caeiro Laboratory of Parasitology, MED-Mediterranean Institute for Agriculture, Environment and Development, Department of Veterinary Medicine, IIFA, University of Évora, Pólo da Mitra, Évora, Portugal
| |
Collapse
|
18
|
Metwali A, Winckler S, Urban JF, Kaplan MH, Ince MN, Elliott DE. Helminth-induced regulation of T-cell transfer colitis requires intact and regulated T cell Stat6 signaling in mice. Eur J Immunol 2020; 51:433-444. [PMID: 33067820 DOI: 10.1002/eji.201848072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 07/24/2020] [Indexed: 02/01/2023]
Abstract
Infection with parasitic worms (helminths) alters host immune responses and can inhibit pathogenic inflammation. Helminth infection promotes a strong Th2 and T regulatory response while suppressing Th1 and Th17 function. Th2 responses are largely dependent on transcriptional programs directed by Stat6-signaling. We examined the importance of intact T cell Stat6 signaling on helminth-induced suppression of murine colitis that results from T cell transfer into immune-deficient mice. Colonization with the intestinal nematode Heligmosomoides polygyrus bakeri resolves WT T cell transfer colitis. However, if the transferred T cells lack intact Stat6 then helminth exposure failed to attenuate colitis or suppress MLN T cell IFN-γ or IL17 production. Loss of Stat6 signaling resulted in decreased IL10 and increased IFN-γ co-expression by IL-17+ T cells. We also transferred T cells from mice with constitutive T cell expression of activated Stat6 (Stat6VT). These mice developed a severe eosinophilic colitis that also was not attenuated by helminth infection. These results show that T cell expression of intact but regulated Stat6 signaling is required for helminth infection-associated regulation of pathogenic intestinal inflammation.
Collapse
Affiliation(s)
- Ahmed Metwali
- Internal Medicine, Iowa City Veterans Administration Health Center, Iowa City, IA, USA.,Department of Internal Medicine, University of Iowa, Carver College of Medicine, Iowa City, IA, USA
| | - Sarah Winckler
- Internal Medicine, Iowa City Veterans Administration Health Center, Iowa City, IA, USA.,Department of Internal Medicine, University of Iowa, Carver College of Medicine, Iowa City, IA, USA
| | - Joseph F Urban
- U.S. Department of Agriculture, Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics, and Immunology Laboratory, Beltsville, MD, USA
| | - Mark H Kaplan
- Department of Pediatrics, H.B. Wells Center for Pediatric Research and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - M Nedim Ince
- Internal Medicine, Iowa City Veterans Administration Health Center, Iowa City, IA, USA.,Department of Internal Medicine, University of Iowa, Carver College of Medicine, Iowa City, IA, USA
| | - David E Elliott
- Internal Medicine, Iowa City Veterans Administration Health Center, Iowa City, IA, USA.,Department of Internal Medicine, University of Iowa, Carver College of Medicine, Iowa City, IA, USA
| |
Collapse
|
19
|
"Begging the Question"-Does Toxocara Infection/Exposure Associate with Multiple Sclerosis-Risk? Pathogens 2020; 9:pathogens9110938. [PMID: 33187271 PMCID: PMC7696196 DOI: 10.3390/pathogens9110938] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 11/01/2020] [Accepted: 11/04/2020] [Indexed: 12/14/2022] Open
Abstract
Although the cause of multiple sclerosis (MS) is unclear, infectious agents, including some parasitic roundworms (nematodes), have been proposed as possible risk factors or contributors. Here, we conducted a systematic review and meta-analysis of published observational studies to evaluate whether there is a possible association between infection with, or exposure to, one or more members of the genus Toxocara (phylum Nematoda; superfamily Ascaridoidea) and MS. We undertook a search of public literature databases to identify relevant studies and then used a random-effects meta-analysis model to generate the pooled odds ratio (OR) and 95% confidence intervals (CIs). This search identified six of a total of 1371 articles that were relevant to the topic; these published studies involved totals of 473 MS patients and 647 control subjects. Anti-Toxocara IgG serum antibodies were detected in 62 MS patients and 37 controls, resulting in respective seroprevalences of 13.1% (95% CI: 8.2-20.3) and 4.8% (95% CI: 2.5-9.2), indicating an association (pooled OR, 3.01; 95% CI: 1.46-6.21). Because of the publication bias identified (six eligible studies), well-designed and -controlled studies are required in the future to rigorously test the hypothesis that Toxocara infection/exposure has an association with MS.
Collapse
|
20
|
Toxoplasma gondii and multiple sclerosis: a population-based case-control study. Sci Rep 2020; 10:18855. [PMID: 33139781 PMCID: PMC7606604 DOI: 10.1038/s41598-020-75830-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 10/09/2020] [Indexed: 01/02/2023] Open
Abstract
According to the hygiene hypothesis, parasites could have a protective role in the development of Multiple Sclerosis (MS). Our aim was to assess the association between presence of anti-Toxoplasma gondii antibodies and MS. MS patients were randomly selected from a population-based incident cohort of MS patients in the city of Catania. Age and sex-matched controls were randomly selected from the general population. Clinical and sociodemographic variables were recorded with a structured questionnaire and a blood sample was taken for serological analysis. Specific T. gondii IgG have been detected with a commercial kit. Adjusted Odds Ratios (ORs) were estimated using unconditional logistic regression. 129 MS subjects (66.7% women with a mean age 44.7 ± 11.0 years) and 287 controls (67.3% women with a mean age 48.1 ± 15.6 years) have been enrolled in the study. Anti-T. gondii antibodies were found in 38 cases (29.5%) and 130 controls (45.4%) giving an adjusted OR of 0.56 (95%CI 0.34–0.93). History of mononucleosis and high educational level were significantly associated with MS (adjOR 2.22 and 1.70 respectively) while an inverse association was found between high educational level and T. gondii seropositivity (adjOR 0.42). Our results further support the protective role of parasitic infections in MS.
Collapse
|
21
|
Koneczny I. Update on IgG4-mediated autoimmune diseases: New insights and new family members. Autoimmun Rev 2020; 19:102646. [PMID: 32801046 DOI: 10.1016/j.autrev.2020.102646] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 03/08/2020] [Indexed: 12/23/2022]
Abstract
Antibodies of IgG4 subclass are exceptional players of the immune system, as they are considered to be immunologically inert and functionally monovalent, and as such may be part of classical tolerance mechanisms. IgG4 antibodies are found in a range of different diseases, including IgG4-related diseases, allergy, cancer, rheumatoid arthritis, helminth infection and IgG4 autoimmune diseases, where they may be pathogenic or protective. IgG4 autoimmune diseases are an emerging new group of diseases that are characterized by pathogenic, antigen-specific autoantibodies of IgG4 subclass, such as MuSK myasthenia gravis, pemphigus vulgaris and thrombotic thrombocytopenic purpura. The list of IgG4 autoantigens is rapidly growing and to date contains 29 candidate antigens. Interestingly, IgG4 autoimmune diseases are restricted to four distinct organs: 1) the central and peripheral nervous system, 2) the kidney, 3) the skin and mucous membranes and 4) the vascular system and soluble antigens in the blood circulation. The pathogenicity of IgG4 can be validated using our classification system, and is usually excerted by functional blocking of protein-protein interaction.
Collapse
Affiliation(s)
- Inga Koneczny
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Währingergürtel 18-20, 1090 Vienna, Austria.
| |
Collapse
|
22
|
Castro Rocha FA, Duarte-Monteiro AM, Henrique da Mota LM, Matias Dinelly Pinto AC, Fonseca JE. Microbes, helminths, and rheumatic diseases. Best Pract Res Clin Rheumatol 2020; 34:101528. [PMID: 32448639 PMCID: PMC7203059 DOI: 10.1016/j.berh.2020.101528] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
There has been a progressive interest on modifications of the human defense system following insults occurring in the interface between our body and the external environment, as they may provoke or worsen disease states. Studies suggest that billions of germs, which compose the gut microbiota influence one's innate and adaptive immune responses at the intestinal level, but these microorganisms may also impact rheumatic diseases. The microbiota of the skin, respiratory, and urinary tracts may also be relevant in rheumatology. Evidence indicates that changes in the gut microbiome alter the pathogenesis of immune-mediated diseases such as rheumatoid arthritis and ankylosing spondylitis but also of other disorders like atherosclerosis and osteoarthritis. Therapeutic strategies to modify the microbiota, including probiotics and fecal microbiota transplantation, have been received with skepticism, which, in turn, has drawn attention back to previously developed interventions such as antibiotics. Helminths adapted to humans over the evolution process, but their role in disease modulation, particularly immune-mediated diseases, remains to be understood. The present review focuses on data concerning modifications of the immune system induced by interactions with microbes and pluricellular organisms, namely helminths, and their impact on rheumatic diseases. Practical aspects, including specific microbiota-targeted therapies, are also discussed.
Collapse
Affiliation(s)
- Francisco Airton Castro Rocha
- Departamento de Medicina Clínica, Liga de Reumatologia e Doenças Autoimunes, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brazil.
| | - Ana Margarida Duarte-Monteiro
- Serviço de Reumatologia e Doenças Ósseas Metabólicas, Hospital de Santa Maria, CHULN and Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisboa, Portugal
| | - Licia Maria Henrique da Mota
- Hospital Universitário de Brasília, Programa de Pós-graduação em Ciências Médicas, Faculdade de Medicina, Universidade de Brasília, Brazil
| | - Ana Carolina Matias Dinelly Pinto
- Departamento de Medicina Clínica, Liga de Reumatologia e Doenças Autoimunes, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - João Eurico Fonseca
- Serviço de Reumatologia e Doenças Ósseas Metabólicas, Hospital de Santa Maria, CHULN and Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisboa, Portugal
| |
Collapse
|
23
|
Dawson HD, Chen C, Li RW, Bell LN, Shea-Donohue T, Kringel H, Beshah E, Hill DE, Urban JF. Molecular and metabolomic changes in the proximal colon of pigs infected with Trichuris suis. Sci Rep 2020; 10:12853. [PMID: 32732949 PMCID: PMC7393168 DOI: 10.1038/s41598-020-69462-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 07/06/2020] [Indexed: 12/14/2022] Open
Abstract
The pig whipworm Trichuris suis is important in swine production because of its negative effects on pig performance and, notably, to some humans with inflammatory bowel disease as a therapeutic agent that modulates inflammation. The proximal colon of T. suis-infected pigs exhibited general inflammation around day 21 after inoculation with infective eggs that is transcriptionally characterized by markers of type-2 immune activation, inflammation, cellular infiltration, tissue repair enzymes, pathways of oxidative stress, and altered intestinal barrier function. Prominent gene pathways involved the Th2-response, de novo cholesterol synthesis, fructose and glucose metabolism, basic amino acid metabolism, and bile acid transport. Upstream regulatory factor analysis implicated the bile acid/farnesoid X receptor in some of these processes. Metabolic analysis indicated changes in fatty acids, antioxidant capacity, biochemicals related to methylation, protein glycosylation, extracellular matrix structure, sugars, Krebs cycle intermediates, microbe-derived metabolites and altered metabolite transport. Close to 1,200 differentially expressed genes were modulated in the proximal colon of pigs with a persistent adult worm infection that was nearly 90% lower in pigs that had expelled worms. The results support a model to test diets that favorably alter the microbiome and improve host intestinal health in both pigs and humans exposed to Trichuris.
Collapse
Affiliation(s)
- Harry D Dawson
- United States Department of Agriculture, Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics, and Immunology Laboratory, Beltsville, USA
| | - Celine Chen
- United States Department of Agriculture, Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics, and Immunology Laboratory, Beltsville, USA
| | - Robert W Li
- Beltsville Agricultural Research Center, Animal Parasitic Diseases Laboratory, Beltsville, MD, USA
| | | | | | - Helene Kringel
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ethiopia Beshah
- United States Department of Agriculture, Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics, and Immunology Laboratory, Beltsville, USA
| | - Dolores E Hill
- Beltsville Agricultural Research Center, Animal Parasitic Diseases Laboratory, Beltsville, MD, USA
| | - Joseph F Urban
- United States Department of Agriculture, Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics, and Immunology Laboratory, Beltsville, USA. .,Beltsville Agricultural Research Center, Animal Parasitic Diseases Laboratory, Beltsville, MD, USA.
| |
Collapse
|
24
|
Chitikina SS, Buddiga P, Deb PK, Mailavaram RP, Venugopala KN, Nair AB, Al-Jaidi B, Kar S. Synthesis and anthelmintic activity of some novel (E)-2-methyl/propyl-4-(2-(substitutedbenzylidene)hydrazinyl)-5,6,7,8-tetrahydrobenzo[4,5]thieno[2,3-d]pyrimidines. Med Chem Res 2020. [DOI: 10.1007/s00044-020-02586-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
25
|
Gao S, Li H, Xie H, Wu S, Yuan Y, Chu L, Sun S, Yang H, Wu L, Bai Y, Zhou Q, Wang X, Zhan B, Cui H, Yang X. Therapeutic efficacy of Schistosoma japonicum cystatin on sepsis-induced cardiomyopathy in a mouse model. Parasit Vectors 2020; 13:260. [PMID: 32423469 PMCID: PMC7236195 DOI: 10.1186/s13071-020-04104-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 04/27/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Myocardial dysfunction is one of the most common complications of multiple organ failure in septic shock and significantly increases mortality in patients with sepsis. Although many studies having confirmed that helminth-derived proteins have strong immunomodulatory functions and could treat inflammatory diseases, there is no report on the therapeutic effect of Schistosoma japonicum-produced cystatin (Sj-Cys) on sepsis-induced cardiac dysfunction. METHODS A model of sepsis-induced myocardial injury was established by cecal ligation and puncture (CLP) in mice. Upon CLP operation, each mouse was intraperitoneally treated with 10 µg of recombinant Sj-Cys (rSj-Cys). Twelve hours after CLP, the systolic and diastolic functions of the left ventricular were examined by echocardiography. The levels of myoglobin (Mb), cardiac troponin I (cTnI), N-terminal pro-Brain Natriuretic peptide (NT-proBNP) in sera, and the activity of myeloperoxidase (MPO) in cardiac tissues were examined as biomarkers for heart injury. The heart tissue was collected for checking pathological changes, macrophages and pro-inflammatory cytokine levels. To address the signaling pathway involved in the anti-inflammatory effects of rSj-Cys, myeloid differentiation factor 88 (MyD88) was determined in heart tissue of mice with sepsis and LPS-stimulated H9C2 cardiomyocytes. In addition, the therapeutic effects of rSj-Cys on LPS-induced cardiomyocyte apoptosis were also detected. The levels of M1 biomarker iNOS and M2 biomarker Arg-1 were detected in heart tissue. The pro-inflammatory cytokines TNF-α and IL-6, and regulatory cytokines IL-10 and TGF-β were measured in sera and their mRNA levels in heart tissue of rSj-Cys-treated mice. RESULTS After rSj-Cys treatment, the sepsis-induced heart malfunction was largely improved. The inflammation and injury of heart tissue were significantly alleviated, characterized as significantly decreased infiltration of inflammatory cells in cardiac tissues and fiber swelling, reduced levels of Mb, cTnI and NT-proBNP in sera, and MPO activity in heart tissue. The therapeutic efficacy of rSj-Cys is associated with downregulated pro-inflammatory cytokines (TNF-α and IL-6) and upregulated regulatory inflammatory cytokines (IL-10 and TGF-β), possibly through inhibiting the LPS-MyD88 signal pathway. CONCLUSIONS RSj-Cys significantly reduced sepsis-induced cardiomyopathy and could be considered as a potential therapeutic agent for the prevention and treatment of sepsis associated cardiac dysfunction.
Collapse
Affiliation(s)
- Shifang Gao
- Second Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China.,Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
| | - Huihui Li
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China.,Basic Medical College of Bengbu Medical College, Bengbu, 233000, China
| | - Hong Xie
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
| | - Shili Wu
- First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
| | - Yuan Yuan
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China.,Basic Medical College of Bengbu Medical College, Bengbu, 233000, China
| | - Liang Chu
- Second Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
| | - Siying Sun
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
| | - Huijuan Yang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
| | - Lingqin Wu
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
| | - Yongsheng Bai
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
| | - Qiao Zhou
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
| | - Xin Wang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
| | - Bin Zhan
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Hu Cui
- Second Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China.
| | - Xiaodi Yang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China. .,Basic Medical College of Bengbu Medical College, Bengbu, 233000, China.
| |
Collapse
|
26
|
Khudhair Z, Alhallaf R, Eichenberger RM, Whan J, Kupz A, Field M, Krause L, Wilson DT, Daly NL, Giacomin P, Sotillo J, Loukas A. Gastrointestinal Helminth Infection Improves Insulin Sensitivity, Decreases Systemic Inflammation, and Alters the Composition of Gut Microbiota in Distinct Mouse Models of Type 2 Diabetes. Front Endocrinol (Lausanne) 2020; 11:606530. [PMID: 33613446 PMCID: PMC7892786 DOI: 10.3389/fendo.2020.606530] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 12/18/2020] [Indexed: 12/11/2022] Open
Abstract
Type 2 diabetes (T2D) is a major health problem and is considered one of the top 10 diseases leading to death globally. T2D has been widely associated with systemic and local inflammatory responses and with alterations in the gut microbiota. Microorganisms, including parasitic worms and gut microbes have exquisitely co-evolved with their hosts to establish an immunological interaction that is essential for the formation and maintenance of a balanced immune system, including suppression of excessive inflammation. Herein we show that both prophylactic and therapeutic infection of mice with the parasitic hookworm-like nematode, Nippostrongylus brasiliensis, significantly reduced fasting blood glucose, oral glucose tolerance and body weight gain in two different diet-induced mouse models of T2D. Helminth infection was associated with elevated type 2 immune responses including increased eosinophil numbers in the mesenteric lymph nodes, liver and adipose tissues, as well as increased expression of IL-4 and alternatively activated macrophage marker genes in adipose tissue, liver and gut. N. brasiliensis infection was also associated with significant compositional changes in the gut microbiota at both the phylum and order levels. Our findings show that N. brasiliensis infection drives changes in local and systemic immune cell populations, and that these changes are associated with a reduction in systemic and local inflammation and compositional changes in the gut microbiota which cumulatively might be responsible for the improved insulin sensitivity observed in infected mice. Our findings indicate that carefully controlled therapeutic hookworm infection in humans could be a novel approach for treating metabolic syndrome and thereby preventing T2D.
Collapse
Affiliation(s)
- Zainab Khudhair
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Rafid Alhallaf
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Ramon M. Eichenberger
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Jen Whan
- Advanced Analytical Center, James Cook University, Cairns, QLD, Australia
| | - Andreas Kupz
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Matt Field
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | | | - David T. Wilson
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Norelle L. Daly
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Paul Giacomin
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Javier Sotillo
- Parasitology Reference and Research Laboratory, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Madrid, Spain
| | - Alex Loukas
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- *Correspondence: Alex Loukas,
| |
Collapse
|
27
|
Ben-Ami Shor D, Lachnish J, Bashi T, Dahan S, Shemer A, Segal Y, Shovman O, Halpert G, Volkov A, Barshack I, Amital H, Blank M, Shoenfeld Y. Immunomodulation of Murine Chronic DSS-Induced Colitis by Tuftsin-Phosphorylcholine. J Clin Med 2019; 9:E65. [PMID: 31888063 PMCID: PMC7019495 DOI: 10.3390/jcm9010065] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/10/2019] [Accepted: 12/11/2019] [Indexed: 02/06/2023] Open
Abstract
Helminths or their products can immunomodulate the host immune system, and this phenomenon may be applied as the basis of new anti-inflammatory treatments. Previously, we have shown the efficacy of tuftsin-phosphorylcholine (TPC), based on a helminth product, in four animal models of autoimmune diseases: arthritis, colitis, systemic lupus erythematosus, and experimental autoimmune encephalomyelitis. We demonstrated that TPC reduced inflammatory process ex vivo in peripheral blood lymphocytes (PBLs) and in biopsies from giant-cell arteritis. In the present study, we assessed the therapeutic potential of TPC treatment on a chronic colitis murine model. C57BL/6 mice with chronic colitis were treated with TPC after the third cycle of 2% dextran sodium sulfate (DSS). Oral TPC treatment resulted in amelioration of the colitis clinical manifestations exemplified by reduced disease activity index (DAI) score, expansion of mesenteric lymph nodes (MLN) T regulatory cells (shown by Fluorescence Activated Cell Sorting (FACS)), significant reduction in the expression of pro-inflammatory cytokines (IL-1β, IL17, IL-6, TNFα), and elevation in the expression of anti-inflammatory cytokine IL-10 (shown by RT-PCR). This study demonstrated the potential immunomodulatory effects of oral administration of TPC in a chronic colitis murine model. Further clinical trials are needed in order to evaluate this novel approach for the treatment of patients with inflammatory bowel disease.
Collapse
Affiliation(s)
- Dana Ben-Ami Shor
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center Tel-Hashomer, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 52620, Israel; (J.L.); (T.B.); (S.D.); (A.S.); (Y.S.); (O.S.); (G.H.); (H.A.); (M.B.); (Y.S.)
- Department of Gastroenterology, Tel-Aviv Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6423906, Israel
| | - Jordan Lachnish
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center Tel-Hashomer, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 52620, Israel; (J.L.); (T.B.); (S.D.); (A.S.); (Y.S.); (O.S.); (G.H.); (H.A.); (M.B.); (Y.S.)
| | - Tomer Bashi
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center Tel-Hashomer, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 52620, Israel; (J.L.); (T.B.); (S.D.); (A.S.); (Y.S.); (O.S.); (G.H.); (H.A.); (M.B.); (Y.S.)
| | - Shani Dahan
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center Tel-Hashomer, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 52620, Israel; (J.L.); (T.B.); (S.D.); (A.S.); (Y.S.); (O.S.); (G.H.); (H.A.); (M.B.); (Y.S.)
| | - Asaf Shemer
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center Tel-Hashomer, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 52620, Israel; (J.L.); (T.B.); (S.D.); (A.S.); (Y.S.); (O.S.); (G.H.); (H.A.); (M.B.); (Y.S.)
| | - Yahel Segal
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center Tel-Hashomer, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 52620, Israel; (J.L.); (T.B.); (S.D.); (A.S.); (Y.S.); (O.S.); (G.H.); (H.A.); (M.B.); (Y.S.)
| | - Ora Shovman
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center Tel-Hashomer, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 52620, Israel; (J.L.); (T.B.); (S.D.); (A.S.); (Y.S.); (O.S.); (G.H.); (H.A.); (M.B.); (Y.S.)
| | - Gilad Halpert
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center Tel-Hashomer, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 52620, Israel; (J.L.); (T.B.); (S.D.); (A.S.); (Y.S.); (O.S.); (G.H.); (H.A.); (M.B.); (Y.S.)
| | - Alexander Volkov
- Institute of Pathology, Sheba Medical Center Tel Hashomer, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 52620, Israel; (A.V.); (I.B.)
| | - Iris Barshack
- Institute of Pathology, Sheba Medical Center Tel Hashomer, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 52620, Israel; (A.V.); (I.B.)
| | - Howard Amital
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center Tel-Hashomer, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 52620, Israel; (J.L.); (T.B.); (S.D.); (A.S.); (Y.S.); (O.S.); (G.H.); (H.A.); (M.B.); (Y.S.)
| | - Miri Blank
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center Tel-Hashomer, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 52620, Israel; (J.L.); (T.B.); (S.D.); (A.S.); (Y.S.); (O.S.); (G.H.); (H.A.); (M.B.); (Y.S.)
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center Tel-Hashomer, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 52620, Israel; (J.L.); (T.B.); (S.D.); (A.S.); (Y.S.); (O.S.); (G.H.); (H.A.); (M.B.); (Y.S.)
| |
Collapse
|
28
|
Lee SC, Tang MS, Easton AV, Devlin JC, Chua LL, Cho I, Moy FM, Khang TF, Lim YAL, Loke P. Linking the effects of helminth infection, diet and the gut microbiota with human whole-blood signatures. PLoS Pathog 2019; 15:e1008066. [PMID: 31841569 PMCID: PMC6913942 DOI: 10.1371/journal.ppat.1008066] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 09/03/2019] [Indexed: 12/24/2022] Open
Abstract
Helminth infection and dietary intake can affect the intestinal microbiota, as well as the immune system. Here we analyzed the relationship between fecal microbiota and blood profiles of indigenous Malaysians, referred to locally as Orang Asli, in comparison to urban participants from the capital city of Malaysia, Kuala Lumpur. We found that helminth infections had a larger effect on gut microbial composition than did dietary intake or blood profiles. Trichuris trichiura infection intensity also had the strongest association with blood transcriptional profiles. By characterizing paired longitudinal samples collected before and after deworming treatment, we determined that changes in serum zinc and iron levels among the Orang Asli were driven by changes in helminth infection status, independent of dietary metal intake. Serum zinc and iron levels were associated with changes in the abundance of several microbial taxa. Hence, there is considerable interplay between helminths, micronutrients and the microbiota on the regulation of immune responses in humans.
Collapse
Affiliation(s)
- Soo Ching Lee
- Department of Parasitology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- Centre of Excellence for Research in AIDS (CERiA), University of Malaya, Kuala Lumpur, Malaysia
- * E-mail: (SCL); (YALL); (PL)
| | - Mei San Tang
- Department of Microbiology, New York University School of Medicine, New York, New York, United States of America
| | - Alice V. Easton
- Department of Microbiology, New York University School of Medicine, New York, New York, United States of America
| | - Joseph Cooper Devlin
- Department of Microbiology, New York University School of Medicine, New York, New York, United States of America
| | - Ling Ling Chua
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Malaya Medical Centre, Kuala Lumpur, Malaysia
- Department of Paediatrics, Faculty of Medicine, University of Malaya Medical Centre, Kuala Lumpur, Malaysia
| | - Ilseung Cho
- Department of Medicine, Division of Gastroenterology, New York University School of Medicine, New York, New York, United States of America
| | - Foong Ming Moy
- Department of Social and Preventive Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Tsung Fei Khang
- University of Malaya Centre for Data Analytics, University of Malaya, Kuala Lumpur, Malaysia
- Institute of Mathematical Sciences, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| | - Yvonne A. L. Lim
- Department of Parasitology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- Centre of Excellence for Research in AIDS (CERiA), University of Malaya, Kuala Lumpur, Malaysia
- * E-mail: (SCL); (YALL); (PL)
| | - P’ng Loke
- Department of Microbiology, New York University School of Medicine, New York, New York, United States of America
- * E-mail: (SCL); (YALL); (PL)
| |
Collapse
|
29
|
Immunomodulatory effect of Syphacia obvelata in treatment of experimental DSS-induced colitis in mouse model. Sci Rep 2019; 9:19127. [PMID: 31836772 PMCID: PMC6911064 DOI: 10.1038/s41598-019-55552-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 11/29/2019] [Indexed: 02/08/2023] Open
Abstract
The ability of helminth parasite infections to manipulate the immune system of their host towards T regulatory responses has been proposed to suppress the inflammatory response. The aim of this study was to investigate the protective and therapeutic effect of Syphacia obvelata in the treatment of experimental DSS -induced colitis. 50 male C57BL/6 mice were divided into 5 groups: healthy uninfected controls, DSS colitis, receiving only S. obv, preventive (S. obv + DSS) and therapeutic group (DSS + S.obv). Colitis intensity was investigated by measuring body weight changes, stool consistency/bleeding and colon length. To evaluate the immune responses induced by this nematode, TNF-α, IL-10, IL-17, IFN-γ and expressing of FoxP3+ T cells were measured in mesenteric lymph nodes and Peyer’s patches cells. Mice in preventive and therapeutic groups treated with S. obv egg significantly ameliorated the severity of the DSS colitis, indicated by the reduced disease manifestations, improved histopathological scores correlated with the up regulation of Treg responses and down regulation of proinflammatory cytokines. S. obv can prevention and reverse on-going murine DSS colitis. The data suggest that induction of Tregs and change in cytokine profiles during helminthic therapies were responsible for reversed inflammatory events in IBD.
Collapse
|
30
|
Xu YW, Xing RX, Zhang WH, Li L, Wu Y, Hu J, Wang C, Luo QL, Shen JL, Chen X. Toxoplasma ROP16 I/III ameliorated inflammatory bowel diseases via inducing M2 phenotype of macrophages. World J Gastroenterol 2019; 25:6634-6652. [PMID: 31832003 PMCID: PMC6906210 DOI: 10.3748/wjg.v25.i45.6634] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 10/03/2019] [Accepted: 11/13/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is characterized by chronic and non-specific inflammation of the intestinal mucosa and mainly includes ulcerative colitis and Crohn's disease.
AIM To explore the beneficial effect of ToxoROP16I/III-induced M2 phynotype macrophages in homeostasis of IBDs through downregulation of M1 inflammatory cells.
METHODS RAW264.7 macrophages stimulated by lipopolysaccharide (LPS) (M1 cells) were co-cultured with Caco-2 cells as an inflammatory model of IBD in vitro. The expression of ToxoROP16I/III was observed in RAW264.7 macrophages that were transfected with pEGFP-rop16I/III. The phenotypes of M2 and M1 macrophage cells were assessed by quantitative real-time reverse transcriptase polymerase chain reaction and the expression of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, transforming growth factor (TGF)-β1, IL-10, inducible nitric oxide synthase (iNOS), and arginase-1 (Arg-1) was detected. The expression of iNOS, Arg-1, signal transducer and activator of transcription 3 (Stat3), p-Stat3, Stat6, p-Stat6, programmed death ligand-2 (PD-L2), caspase-3, -8, and -9 was analyzed by Western blotting, and Griess assays were performed to detect nitric oxide (NO). TNF-α, IL-1β, IL-6, TGF-β1, and IL-10 expression in the supernatants was detected by enzyme-linked immunosorbent assay, and Caco-2 cell apoptosis was determined by flow cytometry after mixing M1 cells with M2 cells in a Caco-2 cell co-culture system.
RESULTS M1 cells exhibited significantly increased production of iNOS, NO, TNF-α, IL-1β, and IL-6, while ToxoROP16I/III induced macrophage bias to M2 cells in vitro, showing increased expression of Arg-1, IL-10 and TGF-β1 and elevated production of p-Stat3 and p-Stat6. The mixed M1 and M2 cell culture induced by ToxoROP16I/III exhibited decreased production of NO and iNOS and upregulated expression of Arg-1 and PD-L2. Accordingly, Caco-2 cells became apoptotic, and apoptosis-associated proteins such as caspase-3, -8 and -9 were dampened during co-culture of M1 and M2 cells. Flow cytometry analysis showed that co-culture of M1 cells with Caco-2 cells facilitated the apoptosis of Caco-2 cells, but co-culture of M1 and M2 cells alleviated Caco-2 cell apoptosis.
CONCLUSION ToxoROP16I/III-induced M2 macrophages inhibited apoptosis of Caco-2 cells caused by M1 macrophages. This finding may help gain a better understanding of the underlying mechanism and represent a promising therapeutic strategy for IBDs.
Collapse
Affiliation(s)
- Yong-Wei Xu
- Department of Gastroenterology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China
| | - Rui-Xin Xing
- Department of Gastroenterology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China
| | - Wen-Hui Zhang
- Department of Gastroenterology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China
| | - Lu Li
- Department of Gastroenterology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China
| | - Yi Wu
- Department of Gastroenterology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China
| | - Jing Hu
- Department of Gastroenterology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China
| | - Cong Wang
- Department of Pathogen Biology, Provincial Laboratory of Pathogen Biology and Zoonoses Anhui, Anhui Medical University, Hefei 230032, Anhui Province, China
| | - Qing-Li Luo
- Department of Pathogen Biology, Provincial Laboratory of Pathogen Biology and Zoonoses Anhui, Anhui Medical University, Hefei 230032, Anhui Province, China
| | - Ji-Long Shen
- Department of Pathogen Biology, Provincial Laboratory of Pathogen Biology and Zoonoses Anhui, Anhui Medical University, Hefei 230032, Anhui Province, China
| | - Xi Chen
- Department of Gastroenterology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China
| |
Collapse
|
31
|
Anthelmintic and antimicrobial activity of Indigofera oblongifolia leaf extracts. Saudi J Biol Sci 2019; 27:594-598. [PMID: 32210676 PMCID: PMC6997872 DOI: 10.1016/j.sjbs.2019.11.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 11/24/2019] [Accepted: 11/26/2019] [Indexed: 01/11/2023] Open
Abstract
Due to the endless emergence of drug resistant pathogens, there is a constant need for new therapeutic agents for clinical use. The identification of active components in natural products and determining the efficacy of these active components has become the current focus of pharmacological research. The present study aimed to evaluate the anthelmintic and antimicrobial activities of Indigofera oblongifolia leaf extract (ILE) against the earthworm Allolobophora caliginosa, the gram-positive bacteria (Bacillus cereus, Streptococcus pneumoniae, and Staphylococcus aureus), the gram-negative bacteria (Klebsiella pneumoniae, Pseudomonas aeruginosa and Escherichia coli) and the yeast Candida albicans. Methanolic extract of I. oblongifolia leaf was obtained and the total phenolics and flavonoids in ILE were determined. The anthelmintic study was carried out to determine the time to paralysis and time to death of worms using three doses (100, 200, and 300 mg/mL) of ILE. Also, Kirby-Bauer disk diffusion susceptibility method was used to determine the antimicrobial activity of ILE. The results showed that ILE induces paralysis and death of A. caliginosa at all concentration tested faster than the reference drug, Albendazole. Additionally, ILE exhibited prominent antimicrobial activity against all gram-positive bacteria tested but almost no significant activity against the gram-negative bacteria, except K. pneumoniae. ILE showed close similarity to the spectrum of chloramphenicol and cefoxitin activities. Furthermore, C. albicans was highly susceptible to the leaf extracts. Our results showed that ILE is an effective anthelmintic and antimicrobial agent.
Collapse
|
32
|
Bergstrom BJ, Rose RK, Bellows AS. Stomach nematodes of cotton rats: parasites, commensals, or mutualists? J Mammal 2019. [DOI: 10.1093/jmammal/gyz136] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Abstract
We related presence and burden of stomach nematodes to body mass and reproductive allocation in hispid cotton rats (Sigmodon hispidus) from two long-running field studies in Virginia (1983–1984, n = 286; and 1988–1990, n = 425) and one from Georgia 1987–1989 (n = 459). Eighty percent of rats from the earlier Virginia sample were infected, with mean nematode mass of 1,311 mg. In the later samples, 23% (Virginia) and 33% (Georgia) were infected with mean nematode mass of 493 and 769 mg, respectively. Presence of nematodes was positively correlated with host body length for each sex in each sample. We used analysis of covariance to examine length-adjusted residuals for presence of nematodes and mass of nematodes for association with somatic and reproductive response variables. Both body and reproductive masses were either positively associated or not related to nematode presence in the two low-prevalence samples, and either negatively associated or not related to nematode presence in the high-prevalence sample. No relationships were detected between host mass and nematode mass per host in either sex in any sample. There was no effect of nematode presence on litter size of pregnant females, but there was a positive effect of nematode mass on litter size in Georgia. Recent theory provides several possible explanations for such neutral-to-positive effects of stomach nematodes on host fitness, including the evolution of host tolerance to the parasites, fecundity compensation by the hosts, and positive effects on host health via immune modulation.
Collapse
Affiliation(s)
| | - Robert K Rose
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA
| | - A Scott Bellows
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA
| |
Collapse
|
33
|
Genetic evolution and codon usage analysis of NKX-2.5 gene governing heart development in some mammals. Genomics 2019; 112:1319-1329. [PMID: 31377427 DOI: 10.1016/j.ygeno.2019.07.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 07/26/2019] [Accepted: 07/31/2019] [Indexed: 11/21/2022]
Abstract
NKX-2.5 gene is responsible for cardiac development and its targeted disruption apprehends cardiac development at the linear heart tube stage. Bioinformatic analysis was employed to investigate the codon usage pattern and dN/dS of mammalian NKX-2.5 gene. The relative synonymous codon usage analysis revealed variation in codon usage and two synonymous codons namely ATA (Ile) and GTA (Val) were absent in NKX-2.5 gene across selected mammalian species suggesting that these two codons were possibly selected against during evolution. Parity rule 2 analysis of two and four fold amino acids showed CT bias whereas six-fold amino acids revealed GA bias. Neutrality analysis suggests that selection played a prominent role while mutation had a minor role. The dN/dS analysis suggests synonymous substitution played a significant role and it negatively correlated with p-distance of the gene. Purifying natural selection played a dominant role in the genetic evolution of NKX-2.5 gene in mammals.
Collapse
|
34
|
Ali Mubaraki M, Ahmad M, Hafiz TA, Marie MA. The therapeutic prospect of crosstalk between prokaryotic and eukaryotic organisms in the human gut. FEMS Microbiol Ecol 2019; 94:4966977. [PMID: 29796663 DOI: 10.1093/femsec/fiy065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 04/09/2018] [Indexed: 12/19/2022] Open
Abstract
The peaceful phenomenon of the co-evolution between the prokaryotes (microbiota) and the eukaryotes (parasites including protozoa and helminths) in the animal gut has drawn the researchers' attention. Importantly, exploring the potential of helminths for therapeutic uses was one of the reasons behind understanding the physiological and immunological crosstalk existing between them. Here we discuss the interactive immunological associations of helminths and microbial responses individually and in combination with their hosts. Considering that there is probably crosstalk between eukaryotic organisms like helminths and protozoa with their host's gut microbiota, in this review we searched the literature identifying the privileged and favourable relationship generated between them in the host. Understanding the possibilities of the role of helminths along with gut microbiota as a black box would certainly help decode the therapeutic intrusion with helminths in experimental clinical trials, and a successful trial could be used to consider possible future and safe treatments for various immune-inflammatory diseases in humans.
Collapse
Affiliation(s)
- Murad Ali Mubaraki
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, King Saud University, Saudi Arabia
| | - Mohammad Ahmad
- Medical Surgical Nursing Department, College of Nursing, King Saud University, Saudi Arabia
| | - Taghreed A Hafiz
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, King Saud University, Saudi Arabia
| | - Mohammed A Marie
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, King Saud University, Saudi Arabia
| |
Collapse
|
35
|
Kuehn C, Tauchi M, Furtmair R, Urschel K, Raaz-Schrauder D, Neumann AL, Frohberger SJ, Hoerauf A, Regus S, Lang W, Sagban TA, Stumpfe FM, Achenbach S, Hübner MP, Dietel B. Filarial extract of Litomosoides sigmodontis induces a type 2 immune response and attenuates plaque development in hyperlipidemic ApoE-knockout mice. FASEB J 2019; 33:6497-6513. [PMID: 30807258 DOI: 10.1096/fj.201800947rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A type 1 immune response is involved in atherosclerosis progression, whereas the role of a type 2 polarization, especially with regard to an enhanced T helper (Th)2 cell differentiation, is still unclear. Helminths trigger type 2 immune responses, protecting the host from inflammatory disorders. We investigated whether an increased type 2 polarization by administration of Litomosoides sigmodontis adult worm extract (LsAg) affects atherosclerosis in apolipoprotein E-deficient (ApoE-/-) mice. Injections of 50 µg LsAg, i.p. into ApoE-/- mice induced a type 2 immune response shown by increased frequencies of peritoneal eosinophils and alternatively activated macrophages. To analyze the effect of LsAg on atherosclerosis initiation, ApoE-/- mice received a high-fat diet for 12 wk and weekly injections of 50 µg LsAg from wk 5 to 12. Therapeutic effects on advanced atherosclerosis were analyzed in mice that were fed a high-fat diet for 12 wk followed by 12 wk of normal chow and weekly LsAg injections. Both preventive and therapeutic LsAg application significantly decreased plaque size. Therapeutic treatment even caused regression of plaque size and macrophage density in the aortic root and reduced Th1-specific gene expression and intraplaque inflammation. In addition, plaque size after therapeutic treatment was inversely correlated with plaque-infiltrated alternatively activated macrophages. In vitro, LsAg treatment of HUVECs reduced intracellular levels of phosphorylated NF-κB-p65, IκB-α, and JNK1/2. In bifurcation flow-through slides, THP-1 cell adhesion to a HUVEC monolayer was decreased by LsAg in regions of nonuniform shear stress. Applying inhibitors of the respective kinases suggests JNK1/2 inhibition is involved in the suppressed cell adhesion. A switch to an enhanced type 2 immune response by LsAg exerts antiatherogenic effects on murine plaque development, indicating a protective role of a hampered type 1 polarization. In vitro, LsAg affects endothelial signaling pathways, among which JNK1/2 inhibition seems to be involved in the suppression of monocytic cell adhesion under proatherogenic shear stress.-Constanze, K., Tauchi, M., Furtmair, R., Urschel, K., Raaz-Schrauder, D., Neumann, A.-L., Frohberger, S. J., Hoerauf, A., Regus, S., Lang, W., Sagban, T. A., Stumpfe, F. M., Achenbach, S., Hübner, M. P., Dietel, B. Filarial extract of Litomosoides sigmodontis induces a type 2 immune response and attenuates plaque development in hyperlipidemic ApoE-knockout mice.
Collapse
Affiliation(s)
- Constanze Kuehn
- Department of Medicine 2-Cardiology and Angiology, Friedrich-Alexander-University (FAU) Erlangen-Nuernberg, Erlangen, Germany
| | - Miyuki Tauchi
- Department of Medicine 2-Cardiology and Angiology, Friedrich-Alexander-University (FAU) Erlangen-Nuernberg, Erlangen, Germany
| | - Roman Furtmair
- Department of Medicine 2-Cardiology and Angiology, Friedrich-Alexander-University (FAU) Erlangen-Nuernberg, Erlangen, Germany
| | - Katharina Urschel
- Department of Medicine 2-Cardiology and Angiology, Friedrich-Alexander-University (FAU) Erlangen-Nuernberg, Erlangen, Germany
| | - Dorette Raaz-Schrauder
- Department of Medicine 2-Cardiology and Angiology, Friedrich-Alexander-University (FAU) Erlangen-Nuernberg, Erlangen, Germany
| | - Anna-Lena Neumann
- Institute for Medical Microbiology, Immunology, and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Stefan J Frohberger
- Institute for Medical Microbiology, Immunology, and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Achim Hoerauf
- Institute for Medical Microbiology, Immunology, and Parasitology, University Hospital Bonn, Bonn, Germany.,German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| | - Susanne Regus
- Department of Vascular Surgery, University Hospital Erlangen, Erlangen, Germany Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Werner Lang
- Department of Vascular Surgery, University Hospital Erlangen, Erlangen, Germany Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Tolga Atilla Sagban
- Department of Vascular Surgery, University Hospital Erlangen, Erlangen, Germany Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany.,Sana-Klinikum Hameln-Pyrmont, Hameln, Germany
| | | | - Stephan Achenbach
- Department of Medicine 2-Cardiology and Angiology, Friedrich-Alexander-University (FAU) Erlangen-Nuernberg, Erlangen, Germany
| | - Marc P Hübner
- Institute for Medical Microbiology, Immunology, and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Barbara Dietel
- Department of Medicine 2-Cardiology and Angiology, Friedrich-Alexander-University (FAU) Erlangen-Nuernberg, Erlangen, Germany
| |
Collapse
|
36
|
Aguayo V, Valdés Fernandez BN, Rodríguez-Valentín M, Ruiz-Jiménez C, Ramos-Benítez MJ, Méndez LB, Espino AM. Fasciola hepatica GST downregulates NF-κB pathway effectors and inflammatory cytokines while promoting survival in a mouse septic shock model. Sci Rep 2019; 9:2275. [PMID: 30783117 PMCID: PMC6381083 DOI: 10.1038/s41598-018-37652-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/28/2018] [Indexed: 12/14/2022] Open
Abstract
Parasitic helminths and helminth-derived molecules have demonstrated to possess powerful anti-inflammatory properties and confirmed therapeutic effects on inflammatory diseases. The helminth Fasciola hepatica has been reported to suppress specific Th1 specific immune responses induced by concurrent bacterial infections, thus demonstrating its anti-inflammatory ability in vivo. In this study, we demonstrate that native F. hepatica glutathione S-transferase (nFhGST), a major parasite excretory-secretory antigen, majorly comprised of Mu-class GST isoforms, significantly suppresses the LPS-induced TNFα and IL1β of mouse bone-marrow derived macrophages in vitro and the pro-inflammatory cytokine/chemokine storm within C57BL/6 mice exposed to lethal doses of LPS increasing their survival rate by more than 85%. Using THP1-Blue CD14 cells, a human monocyte cell line, we also demonstrate that nFhGST suppresses NF-κB activation in response to multiple TLR-ligands, including whole bacteria clinical isolates and this suppression was found to be dose-dependent and independent of the timing of exposure. Moreover, the suppressive effect of nFhGST on NF-κB activation was shown to be independent of enzyme activity or secondary structure of protein. As part of its anti-inflammatory effect nFhGST target multiple proteins of the canonic and non-canonic NF-κB signaling pathway as well as also JAK/STAT pathway. Overall, our results demonstrate the potent anti-inflammatory properties of nFhGST and its therapeutic potential as an anti-inflammatory agent.
Collapse
Affiliation(s)
- Vasti Aguayo
- University of Puerto Rico, Medical Sciences Campus, Department of Microbiology, San Juan, Puerto Rico
| | | | | | - Caleb Ruiz-Jiménez
- University of Puerto Rico, Medical Sciences Campus, Department of Microbiology, San Juan, Puerto Rico
| | - Marcos J Ramos-Benítez
- University of Puerto Rico, Medical Sciences Campus, Department of Microbiology, San Juan, Puerto Rico
| | - Loyda B Méndez
- School of Science & Technology Universidad del Este, Carolina, Puerto Rico
| | - Ana M Espino
- University of Puerto Rico, Medical Sciences Campus, Department of Microbiology, San Juan, Puerto Rico.
| |
Collapse
|
37
|
Arroyo-López C. Helminth therapy for autism under gut-brain axis- hypothesis. Med Hypotheses 2019; 125:110-118. [PMID: 30902137 DOI: 10.1016/j.mehy.2019.02.042] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 02/18/2019] [Indexed: 12/20/2022]
Abstract
Autism is a neurodevelopmental disease included within Autism Syndrome Disorder (ASD) spectrum. ASD has been linked to a series of genes that play a role in immune response function and patients with autism, commonly suffer from immune-related comorbidities. Despite the complex pathophysiology of autism, Gut-brain axis is gaining strength in the understanding of several neurological disorders. In addition, recent publications have shown the correlation between immune dysfunctions, gut microbiota and brain with the behavioral alterations and comorbid symptoms found in autism. Gut-brain axis acts as the "second brain", in a communication network established between neural, endocrine and the immunological systems. On the other hand, Hygiene Hypothesis suggests that the increase in the incidence of autoimmune diseases in the modern world can be attributed to the decrease of exposure to infectious agents, as parasitic nematodes. Helminths induce modulatory and protective effects against several inflammatory disorders, maintaining gastrointestinal homeostasis and modulating brain functions. Helminthic therapy has been previously performed in diseases such as ulcerative colitis, Crohn's disease, diabetes, multiple sclerosis, asthma, rheumatoid arthritis, and food allergies. Considering gut-brain axis, Hygiene Hypothesis, and the modulatory effects of helminths I hypothesized that a treatment with Trichuris suis soluble products represents a feasible holistic treatment for autism, and the key for the development of novel treatments. Preclinical studies are required to test this hypothesis.
Collapse
Affiliation(s)
- Celia Arroyo-López
- Department of Pathology and Laboratory Medicine, UC Davis School of Medicine; Institute for Pediatric Regenerative Medicine and Shriners Hospitals for Children of Northern California, United States.
| |
Collapse
|
38
|
Tang CL, Zou JN, Zhang RH, Liu ZM, Mao CL. Helminths protect against type 1 diabetes: effects and mechanisms. Parasitol Res 2019; 118:1087-1094. [PMID: 30758662 DOI: 10.1007/s00436-019-06247-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 02/01/2019] [Indexed: 02/07/2023]
Abstract
Type 1 diabetes (T1D) is an autoimmune disease in which cells of the immune system destroy pancreatic β cells, which secrete insulin. The high prevalence of T1D in developed societies may be explained by environmental changes, including lower exposure to helminths. Indeed, infection by helminths such as Schistosoma, Filaria, and Heligmosomoides polygyrus and their by-products has been reported to ameliorate or prevent the development of T1D in human and animal models. Helminths can trigger distinct immune regulatory pathways, often involving adaptive immune cells that include T helper 2 (Th2) cells and regulatory T cells (Tregs) and innate immune cells that include dendritic cells, macrophages, and invariant natural killer T cells, which may act synergistically to induce Tregs in a Toll-like receptor-dependent manner. Cytokines such as interleukin (IL)-4, IL-10, and transforming growth factor (TGF)-β also play an important role in protection from T1D. Herein, we provide a comprehensive review of the effects and mechanisms underlying protection against T1D by helminths.
Collapse
Affiliation(s)
- Chun-Lian Tang
- Wuchang Hospital affiliated to Wuhan University of Science and Technology, Wuhan, 430063, China
| | - Jie-Ning Zou
- Wuchang Hospital affiliated to Wuhan University of Science and Technology, Wuhan, 430063, China
| | - Rong-Hui Zhang
- Wuchang Hospital affiliated to Wuhan University of Science and Technology, Wuhan, 430063, China
| | - Zhi-Ming Liu
- Wuchang Hospital affiliated to Wuhan University of Science and Technology, Wuhan, 430063, China.
| | - Cun-Lan Mao
- Department of Obstetrics and Gynecology, People's Hospital of Songzi City, Songzi, 434200, Hubei, China.
| |
Collapse
|
39
|
de Miranda LHM, Meli M, Conceição-Silva F, Novacco M, Menezes RC, Pereira SA, Sugiarto S, dos Reis ÉG, Gremião IDF, Hofmann-Lehmann R. Co-infection with feline retrovirus is related to changes in immunological parameters of cats with sporotrichosis. PLoS One 2018; 13:e0207644. [PMID: 30500849 PMCID: PMC6267967 DOI: 10.1371/journal.pone.0207644] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 11/04/2018] [Indexed: 02/03/2023] Open
Abstract
Feline sporotrichosis due to Sporothrix brasiliensis is frequently severe and often correlated to zoonotic transmission. Feline Immunodeficiency Virus (FIV) and Feline Leukemia Virus (FeLV) cause immunodeficiency in cats; no association has been identified with critical cases of sporotrichosis. Moreover, the cytokine profile in Sporothrix-infected cats and a potential impact of retrovirus co-infections on their immunity is unknown. This study assessed immunological parameters in cats with sporotrichosis with and without FIV or FeLV co-infection. FeLV infection was detected by antigen ELISA and by provirus PCR. FIV infection was investigated through ELISA and Western blot. Cytokine transcription (IFN-γ, IL-4, IL-5, IL-6, IL-10, IL-12, TNF-α) was quantified using RT-qPCR and lymphocyte subpopulations (CD4, CD8, CD5 and CD21) were assessed by flow cytometry. Thirty cats with sporotrichosis were recruited to the study, including three FIV-positive and five FeLV-positive (progressive infection) cats. One cat with regressive FeLV infection was excluded from statistics. In comparison to retrovirus-negative cats, FIV-positive cats and FeLV-positive cats had higher IL-10 levels, FeLV-positive cats had lower IL-4 levels and FIV-positive cats had lower IL-12 levels and a lower CD4+/CD8+ ratio. Remarkably, all cats with poor general condition were FeLV (progressive infection) or FIV-positive, but the retrovirus status was not associated with the sporotrichosis treatment length or outcome. The immunological changes and the more severe clinical presentation observed in cats with retrovirus co-infections encourage future prospective studies that address the impact of these changes on prognostic determinants of feline sporotrichosis and the development of new therapy strategies that control disease spread.
Collapse
Affiliation(s)
- Luisa Helena Monteiro de Miranda
- Laboratory of Clinical Research on Dermatozoonoses in Domestic Animals, Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Clinical Laboratory and Center for Clinical Studies, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
- * E-mail:
| | - Marina Meli
- Clinical Laboratory and Center for Clinical Studies, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Fátima Conceição-Silva
- Laboratory of Immunoparasitology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Marilisa Novacco
- Clinical Laboratory and Center for Clinical Studies, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Rodrigo Caldas Menezes
- Laboratory of Clinical Research on Dermatozoonoses in Domestic Animals, Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Sandro Antonio Pereira
- Laboratory of Clinical Research on Dermatozoonoses in Domestic Animals, Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Sarah Sugiarto
- Clinical Laboratory and Center for Clinical Studies, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Érica Guerino dos Reis
- Laboratory of Clinical Research on Dermatozoonoses in Domestic Animals, Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Isabella Dib Ferreira Gremião
- Laboratory of Clinical Research on Dermatozoonoses in Domestic Animals, Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Regina Hofmann-Lehmann
- Clinical Laboratory and Center for Clinical Studies, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| |
Collapse
|
40
|
Leroux LP, Nasr M, Valanparambil R, Tam M, Rosa BA, Siciliani E, Hill DE, Zarlenga DS, Jaramillo M, Weinstock JV, Geary TG, Stevenson MM, Urban JF, Mitreva M, Jardim A. Analysis of the Trichuris suis excretory/secretory proteins as a function of life cycle stage and their immunomodulatory properties. Sci Rep 2018; 8:15921. [PMID: 30374177 PMCID: PMC6206011 DOI: 10.1038/s41598-018-34174-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 10/12/2018] [Indexed: 12/21/2022] Open
Abstract
Parasitic worms have a remarkable ability to modulate host immune responses through several mechanisms including excreted/secreted proteins (ESP), yet the exact nature of these proteins and their targets often remains elusive. Here, we performed mass spectrometry analyses of ESP (TsESP) from larval and adult stages of the pig whipworm Trichuris suis (Ts) and identified ~350 proteins. Transcriptomic analyses revealed large subsets of differentially expressed genes in the various life cycle stages of the parasite. Exposure of bone marrow-derived macrophages and dendritic cells to TsESP markedly diminished secretion of the pro-inflammatory cytokines TNFα and IL-12p70. Conversely, TsESP exposure strongly induced release of the anti-inflammatory cytokine IL-10, and also induced high levels of nitric oxide (NO) and upregulated arginase activity in macrophages. Interestingly, TsESP failed to directly induce CD4+ CD25+ FoxP3+ regulatory T cells (Treg cells), while OVA-pulsed TsESP-treated dendritic cells suppressed antigen-specific OT-II CD4+ T cell proliferation. Fractionation of TsESP identified a subset of proteins that promoted anti-inflammatory functions, an activity that was recapitulated using recombinant T. suis triosephosphate isomerase (TPI) and nucleoside diphosphate kinase (NDK). Our study helps illuminate the intricate balance that is characteristic of parasite-host interactions at the immunological interface, and further establishes the principle that specific parasite-derived proteins can modulate immune cell functions.
Collapse
Affiliation(s)
- Louis-Philippe Leroux
- Institute of Parasitology McGill University, Sainte-Anne-de-Bellevue, QC, Canada
- Centre for Host-Parasite Interaction (CHPI), Montreal, Canada
- Institut National de la Recherche Scientifique (INRS)-Institut Armand-Frappier (IAF), Laval, QC, Canada
| | - Mohamad Nasr
- Institute of Parasitology McGill University, Sainte-Anne-de-Bellevue, QC, Canada
- Centre for Host-Parasite Interaction (CHPI), Montreal, Canada
| | - Rajesh Valanparambil
- Centre for Host-Parasite Interaction (CHPI), Montreal, Canada
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, QC, Canada
| | - Mifong Tam
- Centre for Host-Parasite Interaction (CHPI), Montreal, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Bruce A Rosa
- McDonnell Genome Institute, Washington University in, St. Louis, MO, USA
| | - Elizabeth Siciliani
- Institute of Parasitology McGill University, Sainte-Anne-de-Bellevue, QC, Canada
| | - Dolores E Hill
- United States Department of Agriculture, Beltsville, MD, USA
| | | | - Maritza Jaramillo
- Centre for Host-Parasite Interaction (CHPI), Montreal, Canada
- Institut National de la Recherche Scientifique (INRS)-Institut Armand-Frappier (IAF), Laval, QC, Canada
| | - Joel V Weinstock
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, Tufts Medical Center, Boston, MA, USA
| | - Timothy G Geary
- Institute of Parasitology McGill University, Sainte-Anne-de-Bellevue, QC, Canada
- Centre for Host-Parasite Interaction (CHPI), Montreal, Canada
| | - Mary M Stevenson
- Centre for Host-Parasite Interaction (CHPI), Montreal, Canada
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, QC, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Joseph F Urban
- United States Department of Agriculture, Beltsville, MD, USA
| | - Makedonka Mitreva
- McDonnell Genome Institute, Washington University in, St. Louis, MO, USA
- Division of Infectious Diseases, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Armando Jardim
- Institute of Parasitology McGill University, Sainte-Anne-de-Bellevue, QC, Canada.
- Centre for Host-Parasite Interaction (CHPI), Montreal, Canada.
| |
Collapse
|
41
|
Zuo T, Ng SC. The Gut Microbiota in the Pathogenesis and Therapeutics of Inflammatory Bowel Disease. Front Microbiol 2018; 9:2247. [PMID: 30319571 PMCID: PMC6167487 DOI: 10.3389/fmicb.2018.02247] [Citation(s) in RCA: 356] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 09/03/2018] [Indexed: 12/12/2022] Open
Abstract
In the twenty first century, the changing epidemiology of inflammatory bowel disease (IBD) globally with increasing disease incidence across many countries relates to the altered gut microbiota, due to a combinatorial effect of environmental factors, human immune responses and genetics. IBD is a gastrointestinal disease associated with a gut microbial dysbiosis, including an expansion of facultative anaerobic bacteria of the family Enterobacteriaceae. Advances in high-throughput sequencing enable us to entangle the gut microbiota in human health and IBD beyond the gut bacterial microbiota, expanding insights into the mycobiota, virobiota and helminthes. Caudovirales (viruses) and Basidiomycota, Ascomycota, and Candida albicans (fungi) are revealed to be increased in IBD. The deconvolution of the gut microbiota in IBD lays the basis for unveiling the roles of these various gut microbiota components in IBD pathogenesis and being conductive to instructing on future IBD diagnosis and therapeutics. Here we comprehensively elucidate the alterations in the gut microbiota in IBD, discuss the effect of diets in the gut microbiota in relation to IBD, and illustrate the potential of manipulation of gut microbiota for IBD therapeutics. The therapeutic strategy of antibiotics, prebiotics, probiotics and fecal microbiota transplantation will benefit the effective application of precision microbiome manipulation in IBD.
Collapse
Affiliation(s)
- Tao Zuo
- Department of Medicine and Therapeutics, Institute of Digestive Disease, LKS Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
- Faculty of Medicine, Center for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong, China
| | - Siew C. Ng
- Department of Medicine and Therapeutics, Institute of Digestive Disease, LKS Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
- Faculty of Medicine, Center for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
42
|
Czimmerer Z, Daniel B, Horvath A, Rückerl D, Nagy G, Kiss M, Peloquin M, Budai MM, Cuaranta-Monroy I, Simandi Z, Steiner L, Nagy B, Poliska S, Banko C, Bacso Z, Schulman IG, Sauer S, Deleuze JF, Allen JE, Benko S, Nagy L. The Transcription Factor STAT6 Mediates Direct Repression of Inflammatory Enhancers and Limits Activation of Alternatively Polarized Macrophages. Immunity 2018; 48:75-90.e6. [PMID: 29343442 PMCID: PMC5772169 DOI: 10.1016/j.immuni.2017.12.010] [Citation(s) in RCA: 163] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 11/09/2017] [Accepted: 12/11/2017] [Indexed: 11/29/2022]
Abstract
The molecular basis of signal-dependent transcriptional activation has been extensively studied in macrophage polarization, but our understanding remains limited regarding the molecular determinants of repression. Here we show that IL-4-activated STAT6 transcription factor is required for the direct transcriptional repression of a large number of genes during in vitro and in vivo alternative macrophage polarization. Repression results in decreased lineage-determining transcription factor, p300, and RNA polymerase II binding followed by reduced enhancer RNA expression, H3K27 acetylation, and chromatin accessibility. The repressor function of STAT6 is HDAC3 dependent on a subset of IL-4-repressed genes. In addition, STAT6-repressed enhancers show extensive overlap with the NF-κB p65 cistrome and exhibit decreased responsiveness to lipopolysaccharide after IL-4 stimulus on a subset of genes. As a consequence, macrophages exhibit diminished inflammasome activation, decreased IL-1β production, and pyroptosis. Thus, the IL-4-STAT6 signaling pathway establishes an alternative polarization-specific epigenenomic signature resulting in dampened macrophage responsiveness to inflammatory stimuli. IL-4-activated STAT6 acts as a transcriptional repressor in macrophages IL-4-STAT6-repressed enhancers associate with reduced LDTF and p300 binding Inflammatory responsiveness of the IL-4-repressed enhancers is attenuated IL-4 limits the LPS-induced inflammasome activation, IL-1β production, and pyroptosis
Collapse
Affiliation(s)
- Zsolt Czimmerer
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Bence Daniel
- Sanford-Burnham-Prebys Medical Discovery Institute, 6400 Sanger Road, Orlando, FL 32827, USA
| | - Attila Horvath
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Dominik Rückerl
- Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, UK
| | - Gergely Nagy
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; MTA-DE "Lendület" Immunogenomics Research Group, University of Debrecen, Debrecen, Hungary
| | - Mate Kiss
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Matthew Peloquin
- Sanford-Burnham-Prebys Medical Discovery Institute, 6400 Sanger Road, Orlando, FL 32827, USA
| | - Marietta M Budai
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ixchelt Cuaranta-Monroy
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltan Simandi
- Sanford-Burnham-Prebys Medical Discovery Institute, 6400 Sanger Road, Orlando, FL 32827, USA
| | - Laszlo Steiner
- UD-Genomed Medical Genomic Technologies Ltd., Debrecen, Hungary
| | - Bela Nagy
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Szilard Poliska
- Genomic Medicine and Bioinformatic Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Csaba Banko
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsolt Bacso
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ira G Schulman
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Sascha Sauer
- Otto Warburg Laboratory, Max Planck Institute for Molecular Genetics, Berlin, Germany; CU Systems Medicine, University of Würzburg, Würzburg, Germany; Max Delbrück Center for Molecular Medicine (BIMSB and BIH), Berlin, Germany
| | | | - Judith E Allen
- Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, UK
| | - Szilvia Benko
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Laszlo Nagy
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Sanford-Burnham-Prebys Medical Discovery Institute, 6400 Sanger Road, Orlando, FL 32827, USA; MTA-DE "Lendület" Immunogenomics Research Group, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
43
|
Abstract
In this Review, I present evidence supporting a multifactorial causation of childhood acute lymphoblastic leukaemia (ALL), a major subtype of paediatric cancer. ALL evolves in two discrete steps. First, in utero initiation by fusion gene formation or hyperdiploidy generates a covert, pre-leukaemic clone. Second, in a small fraction of these cases, the postnatal acquisition of secondary genetic changes (primarily V(D)J recombination-activating protein (RAG) and activation-induced cytidine deaminase (AID)-driven copy number alterations in the case of ETS translocation variant 6 (ETV6)-runt-related transcription factor 1 (RUNX1)+ ALL) drives conversion to overt leukaemia. Epidemiological and modelling studies endorse a dual role for common infections. Microbial exposures earlier in life are protective but, in their absence, later infections trigger the critical secondary mutations. Risk is further modified by inherited genetics, chance and, probably, diet. Childhood ALL can be viewed as a paradoxical consequence of progress in modern societies, where behavioural changes have restrained early microbial exposure. This engenders an evolutionary mismatch between historical adaptations of the immune system and contemporary lifestyles. Childhood ALL may be a preventable cancer.
Collapse
Affiliation(s)
- Mel Greaves
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK.
| |
Collapse
|
44
|
T-Bet Is Dependent on Stat-4 Inhibiting Acute Colitis but Not Stat-1 Using L4 Somatic Antigen of Heligmosomoides polygyrus. ScientificWorldJournal 2018; 2018:8571920. [PMID: 29977172 PMCID: PMC6011060 DOI: 10.1155/2018/8571920] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 03/25/2018] [Accepted: 04/17/2018] [Indexed: 02/06/2023] Open
Abstract
Helminths may alter the immunoinflammatory reactions of colitis. Proteins derived from H. polygyrus have prospective therapy for colitis. The goal of this study was to interpret the protective mechanisms of L4 somatic antigen (LSA) from Heligmosomoides polygyrus against an inflammatory response to the pathogenesis of DNBS-induced colitis. Colitis was actuated in mice by rectal instillation of DNBS. The mice were randomly divided into five groups containing control, DNBS alone, and three groups, with different doses of LSA (50, 100, and 200 μg/mL), respectively. Mice initiated colitis by rectal administration of DNBS and after that were immunized with LSA for 14 days. Mice treated with LSA inhibited wasting disease compared with DNBS only group. The percentages of cells producing IFN-γ were reduced by LSA treatment. The level of T lymphocytes CD4+IFN-γ+ cells in the LPL was significantly diminished by LSA at both 100 and 200 μg/mL groups (p<0.05). The mRNA expression of T-bet was significantly declined in LSA immunized mice, but not RORγ-T mRNA, whereas GATA-3 expression tended to increase. The activation of STAT-4 significantly reduced LSA-treated mice but not STAT-1. It can be concluded that T-bet is required for optimal production of IFN-γ in colitis.
Collapse
|
45
|
Su CW, Chen CY, Li Y, Long SR, Massey W, Kumar DV, Walker WA, Shi HN. Helminth infection protects against high fat diet-induced obesity via induction of alternatively activated macrophages. Sci Rep 2018; 8:4607. [PMID: 29545532 PMCID: PMC5854586 DOI: 10.1038/s41598-018-22920-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 02/26/2018] [Indexed: 12/11/2022] Open
Abstract
Epidemiological studies indicate an inverse correlation between the prevalence of the so-called western diseases, such as obesity and metabolic syndrome, and the exposure to helminths. Obesity, a key risk factor for many chronic health problems, is rising globally and is accompanied by low-grade inflammation in adipose tissues. The precise mechanism by which helminths modulate metabolic syndrome and obesity is not fully understood. We infected high fat diet (HFD)-induced obese mice with the intestinal nematode parasite Heligmosomoides polygyrus and observed that helminth infection resulted in significantly attenuated obesity. Attenuated obesity corresponded with marked upregulation of uncoupling protein 1 (UCP1), a key protein involved in energy expenditure, in adipose tissue, suppression of glucose and triglyceride levels, and alteration in the expression of key genes involved in lipid metabolism. Moreover, the attenuated obesity in infected mice was associated with enhanced helminth-induced Th2/Treg responses and M2 macrophage polarization. Adoptive transfer of helminth-stimulated M2 cells to mice that were not infected with H. polygyrus resulted in a significant amelioration of HFD-induced obesity and increased adipose tissue browning. Thus, our results provide evidence that the helminth-dependent protection against obesity involves the induction of M2 macrophages.
Collapse
Affiliation(s)
- Chien Wen Su
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, USA
| | - Chih-Yu Chen
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, USA
| | - Yali Li
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, USA
| | - Shao Rong Long
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, USA
| | - William Massey
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, USA
| | - Deepak Vijaya Kumar
- Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, USA
| | - W Allan Walker
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, USA
| | - Hai Ning Shi
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, USA.
| |
Collapse
|
46
|
Rosa BA, Supali T, Gankpala L, Djuardi Y, Sartono E, Zhou Y, Fischer K, Martin J, Tyagi R, Bolay FK, Fischer PU, Yazdanbakhsh M, Mitreva M. Differential human gut microbiome assemblages during soil-transmitted helminth infections in Indonesia and Liberia. MICROBIOME 2018; 6:33. [PMID: 29486796 PMCID: PMC6389212 DOI: 10.1186/s40168-018-0416-5] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 01/26/2018] [Indexed: 05/11/2023]
Abstract
BACKGROUND The human intestine and its microbiota is the most common infection site for soil-transmitted helminths (STHs), which affect the well-being of ~ 1.5 billion people worldwide. The complex cross-kingdom interactions are not well understood. RESULTS A cross-sectional analysis identified conserved microbial signatures positively or negatively associated with STH infections across Liberia and Indonesia, and longitudinal samples analysis from a double-blind randomized trial showed that the gut microbiota responds to deworming but does not transition closer to the uninfected state. The microbiomes of individuals able to self-clear the infection had more alike microbiome assemblages compared to individuals who remained infected. One bacterial taxon (Lachnospiracae) was negatively associated with infection in both countries, and 12 bacterial taxa were significantly associated with STH infection in both countries, including Olsenella (associated with reduced gut inflammation), which also significantly reduced in abundance following clearance of infection. Microbial community gene abundances were also affected by deworming. Functional categories identified as associated with STH infection included arachidonic acid metabolism; arachidonic acid is the precursor for pro-inflammatory leukotrienes that threaten helminth survival, and our findings suggest that some modulation of arachidonic acid activity in the STH-infected gut may occur through the increase of arachidonic acid metabolizing bacteria. CONCLUSIONS For the first time, we identify specific members of the gut microbiome that discriminate between moderately/heavily STH-infected and non-infected states across very diverse geographical regions using two different statistical methods. We also identify microbiome-encoded biological functions associated with the STH infections, which are associated potentially with STH survival strategies, and changes in the host environment. These results provide a novel insight of the cross-kingdom interactions in the human gut ecosystem by unlocking the microbiome assemblages at taxonomic, genetic, and functional levels so that advances towards key mechanistic studies can be made.
Collapse
Affiliation(s)
- Bruce A. Rosa
- McDonnell Genome Institute, Washington University, St. Louis, MO 63108 USA
| | - Taniawati Supali
- Department of Parasitology, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Lincoln Gankpala
- Public Health and Medical Research, National Public Health Institute of Liberia, Charlesville, Liberia
| | - Yenny Djuardi
- Department of Parasitology, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Erliyani Sartono
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Yanjiao Zhou
- Microbial Genomics, The Jackson Laboratory for Genomic Medicine, Farmington, CT USA
| | - Kerstin Fischer
- Department of Medicine, Washington University School of Medicine, St. Louis, MO USA
| | - John Martin
- McDonnell Genome Institute, Washington University, St. Louis, MO 63108 USA
| | - Rahul Tyagi
- McDonnell Genome Institute, Washington University, St. Louis, MO 63108 USA
| | - Fatorma K. Bolay
- Public Health and Medical Research, National Public Health Institute of Liberia, Charlesville, Liberia
| | - Peter U. Fischer
- Department of Medicine, Washington University School of Medicine, St. Louis, MO USA
| | - Maria Yazdanbakhsh
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Makedonka Mitreva
- McDonnell Genome Institute, Washington University, St. Louis, MO 63108 USA
- Department of Medicine, Washington University School of Medicine, St. Louis, MO USA
| |
Collapse
|
47
|
Abstract
The microbiota, which is comprised of the collective of all microbes inhabiting the gut and its effect on the human host in which it resides, has become a growing field of interest. Various parameters of health and disease have been found to be associated with the variation in the human gut microbiome. In recent years, many studies have demonstrated an important role of gut microbes in the development of various illnesses including autoimmune diseases, such as type 1 diabetes, rheumatoid arthritis, and multiple sclerosis. Although the mechanism of the disease involves both genetic and environmental factors, lupus has been found to be affected by the composition of the microbes lining the intestines. Several recent studies have suggested that alterations of the gut microbial composition may be correlated with SLE disease manifestations, while the exact roles of either symbiotic or pathogenic microbes in this disease have yet to be explored. Elucidation of the roles of gut microbes in SLE will shed light on how this autoimmune disorder develops and provide opportunities for improved biomarkers of the disease and the potential to probe new therapies. This new knowledge, along with that enabling alteration in composition of the gut microbiome, via diet modification, antibiotic, and probiotics, may bring forward a new era in the future of lupus treatment.
Collapse
Affiliation(s)
- Nurit Katz-Agranov
- Department of Internal Medicine, The University of Texas Houston, Health Science Center, Houston, TX, USA
| | - Gisele Zandman-Goddard
- Department of Medicine C, Wolfson Medical Center, Holon, Israel. .,Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel.
| |
Collapse
|
48
|
Maślińska M, de Luca F, Sharif K. Tuftsin-phosphorylcholine treatment of autoimmune diseases - a benefit and a message from helminths? Reumatologia 2017; 55:267-268. [PMID: 29491533 PMCID: PMC5825963 DOI: 10.5114/reum.2017.72622] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 12/18/2017] [Indexed: 11/23/2022] Open
Affiliation(s)
- Maria Maślińska
- Early Arthritis Clinic National Institute of Geriatrics, Rheumatology, and Rehabilitation, Warsaw, Poland
| | - Fabrizio de Luca
- School of Medicine, Milan University, Department of Allergy and Immunology, Niguarda Ca’ Granda Metropolitan Hospital, Milan, Italy
| | - Kassem Sharif
- Internal Medicine B, Sheba Medical Centre, Tel-Hashomer, Israel
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Centre, Tel-Hashomer, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
49
|
Navarro S, Pickering DA, Ferreira IB, Jones L, Ryan S, Troy S, Leech A, Hotez PJ, Zhan B, Laha T, Prentice R, Sparwasser T, Croese J, Engwerda CR, Upham JW, Julia V, Giacomin PR, Loukas A. Hookworm recombinant protein promotes regulatory T cell responses that suppress experimental asthma. Sci Transl Med 2017; 8:362ra143. [PMID: 27797959 DOI: 10.1126/scitranslmed.aaf8807] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 09/01/2016] [Indexed: 12/12/2022]
Abstract
In the developed world, declining prevalence of some parasitic infections correlates with increased incidence of allergic and autoimmune disorders. Moreover, experimental human infection with some parasitic worms confers protection against inflammatory diseases in phase 2 clinical trials. Parasitic worms manipulate the immune system by secreting immunoregulatory molecules that offer promise as a novel therapeutic modality for inflammatory diseases. We identify a protein secreted by hookworms, anti-inflammatory protein-2 (AIP-2), that suppressed airway inflammation in a mouse model of asthma, reduced expression of costimulatory markers on human dendritic cells (DCs), and suppressed proliferation ex vivo of T cells from human subjects with house dust mite allergy. In mice, AIP-2 was primarily captured by mesenteric CD103+ DCs and suppression of airway inflammation was dependent on both DCs and Foxp3+ regulatory T cells (Tregs) that originated in the mesenteric lymph nodes (MLNs) and accumulated in distant mucosal sites. Transplantation of MLNs from AIP-2-treated mice into naïve hosts revealed a lymphoid tissue conditioning that promoted Treg induction and long-term maintenance. Our findings indicate that recombinant AIP-2 could serve as a novel curative therapeutic for allergic asthma and potentially other inflammatory diseases.
Collapse
Affiliation(s)
- Severine Navarro
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia.
| | - Darren A Pickering
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Ivana B Ferreira
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Linda Jones
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Stephanie Ryan
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Sally Troy
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Andrew Leech
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | | | - Bin Zhan
- Baylor College of Medicine, Houston, TX 77030, USA
| | - Thewarach Laha
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Roger Prentice
- Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Tim Sparwasser
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - John Croese
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia.,Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | | | - John W Upham
- University of Queensland, Brisbane, Queensland, Australia.,Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Valerie Julia
- CNRS UMR7275, INSERM U1080, Université de Nice Sophia Antipolis, Nice, France
| | - Paul R Giacomin
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Alex Loukas
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia.
| |
Collapse
|
50
|
Marzano V, Mancinelli L, Bracaglia G, Del Chierico F, Vernocchi P, Di Girolamo F, Garrone S, Tchidjou Kuekou H, D’Argenio P, Dallapiccola B, Urbani A, Putignani L. "Omic" investigations of protozoa and worms for a deeper understanding of the human gut "parasitome". PLoS Negl Trop Dis 2017; 11:e0005916. [PMID: 29095820 PMCID: PMC5667730 DOI: 10.1371/journal.pntd.0005916] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The human gut has been continuously exposed to a broad spectrum of intestinal organisms, including viruses, bacteria, fungi, and parasites (protozoa and worms), over millions of years of coevolution, and plays a central role in human health. The modern lifestyles of Western countries, such as the adoption of highly hygienic habits, the extensive use of antimicrobial drugs, and increasing globalisation, have dramatically altered the composition of the gut milieu, especially in terms of its eukaryotic “citizens.” In the past few decades, numerous studies have highlighted the composition and role of human intestinal bacteria in physiological and pathological conditions, while few investigations exist on gut parasites and particularly on their coexistence and interaction with the intestinal microbiota. Studies of the gut “parasitome” through “omic” technologies, such as (meta)genomics, transcriptomics, proteomics, and metabolomics, are herein reviewed to better understand their role in the relationships between intestinal parasites, host, and resident prokaryotes, whether pathogens or commensals. Systems biology–based profiles of the gut “parasitome” under physiological and severe disease conditions can indeed contribute to the control of infectious diseases and offer a new perspective of omics-assisted tropical medicine.
Collapse
Affiliation(s)
- Valeria Marzano
- Human Microbiome Unit, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | - Livia Mancinelli
- Laboratory Medicine, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | - Giorgia Bracaglia
- Laboratory Medicine, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | | | - Pamela Vernocchi
- Human Microbiome Unit, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | | | - Stefano Garrone
- Laboratory Medicine, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | | | - Patrizia D’Argenio
- Pediatric Immuno-infectivology, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Bruno Dallapiccola
- Scientific Directorate, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Andrea Urbani
- Institute of Biochemistry and Biochemical Clinic, Faculty of Medicine and Surgery–Policlinico A. Gemelli, Catholic University of Sacred Heart, Rome, Italy
- Proteomic and Metabonomic Unit, Fondazione Santa Lucia IRCCS, Rome, Italy
| | - Lorenza Putignani
- Human Microbiome Unit, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
- Parasitology Unit, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
- * E-mail:
| |
Collapse
|