1
|
Berbudi A, Khairani S, Tjahjadi AI. Interplay Between Insulin Resistance and Immune Dysregulation in Type 2 Diabetes Mellitus: Implications for Therapeutic Interventions. Immunotargets Ther 2025; 14:359-382. [PMID: 40196377 PMCID: PMC11974557 DOI: 10.2147/itt.s499605] [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: 10/07/2024] [Accepted: 02/20/2025] [Indexed: 04/09/2025] Open
Abstract
Type 2 Diabetes Mellitus (T2DM) is a rapidly growing global health issue characterized by insulin resistance and chronic inflammation. Beyond regulating glucose homeostasis, insulin plays a pivotal role in modulating immune cell function, linking metabolic dysregulation with immune responses. This review examines the intricate relationship between insulin resistance and immune dysfunction in T2DM, focusing on how impaired insulin signaling pathways, particularly PI3K/Akt and MAPK, contribute to immune cell activation, proliferation, and chronic inflammation. Insulin resistance impacts immune cells such as T cells, B cells, macrophages, and neutrophils, leading to an imbalance between pro-inflammatory and anti-inflammatory responses. Elevated pro-inflammatory cytokines (eg, TNF-α, IL-6) and adipokines (eg, leptin, resistin) exacerbate insulin resistance, promoting a vicious cycle of metabolic and immune dysregulation. This interplay contributes to the chronic low-grade inflammation that underlies T2DM pathogenesis, further impairing insulin signaling and glucose metabolism. Restoration of insulin sensitivity is, therefore, a critical step toward correcting immune imbalance in insulin-resistant states like T2DM. Therapeutic approaches that reduce inflammation could also support improvements in insulin sensitivity, addressing both metabolic and immune disturbances simultaneously. The review also explores therapeutic strategies, including insulin therapy, targeting insulin signaling pathways, and lifestyle interventions. Insulin therapy can reduce pro-inflammatory cytokine production and enhance anti-inflammatory responses, although challenges such as potential immune suppression and hyperinsulinemia remain. Targeting key signaling pathways and transcription factors offers promising avenues for modulating immune responses, while lifestyle interventions, such as dietary modifications, physical activity, and weight management, can improve insulin sensitivity and reduce inflammation. By understanding the dual role of insulin in regulating both metabolic and immune functions, this review underscores the importance of addressing immune dysfunction as part of comprehensive T2DM management. Targeting the interconnected pathways of insulin signaling and immune regulation could lead to more effective therapeutic approaches, ultimately improving patient outcomes and reducing disease complications.
Collapse
Affiliation(s)
- Afiat Berbudi
- Department of Biomedical Sciences, Parasitology Division, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
- Research Center for Care and Control of Infectious Diseases (RC3ID), Universitas Padjadjaran, Bandung, Indonesia
- Universitas Padjadjaran Hospital, Sumedang, Indonesia
| | - Shafia Khairani
- Department of Biomedical Sciences, Cell Biology Division, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Adi Imam Tjahjadi
- Research Center for Care and Control of Infectious Diseases (RC3ID), Universitas Padjadjaran, Bandung, Indonesia
- Universitas Padjadjaran Hospital, Sumedang, Indonesia
- Department of Biomedical Sciences, Microbiology Division, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| |
Collapse
|
2
|
Zhu Y, Chen X, Zheng H, Ma Q, Chen K, Li H. Anti-Inflammatory Effects of Helminth-Derived Products: Potential Applications and Challenges in Diabetes Mellitus Management. J Inflamm Res 2024; 17:11789-11812. [PMID: 39749005 PMCID: PMC11694023 DOI: 10.2147/jir.s493374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 12/15/2024] [Indexed: 01/04/2025] Open
Abstract
The global rise in diabetes mellitus (DM), particularly type 2 diabetes (T2D), has become a major public health challenge. According to the "hygiene hypothesis", helminth infections may offer therapeutic benefits for DM. These infections are known to modulate immune responses, reduce inflammation, and improve insulin sensitivity. However, they also carry risks, such as malnutrition, anemia, and intestinal obstruction. Importantly, helminth excretory/secretory products, which include small molecules and proteins, have shown therapeutic potential in treating various inflammatory diseases with minimal side effects. This review explores the anti-inflammatory properties of helminth derivatives and their potential to alleviate chronic inflammation in both type 1 diabetes and T2D, highlighting their promise as future drug candidates. Additionally, it discusses the possible applications of these derivatives in DM management and the challenges involved in translating these findings into clinical practice.
Collapse
Affiliation(s)
- Yunhuan Zhu
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, People’s Republic of China
| | - Xintong Chen
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, People’s Republic of China
| | - Hezheng Zheng
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, People’s Republic of China
| | - Qiman Ma
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, People’s Republic of China
| | - Keda Chen
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, People’s Republic of China
| | - Hongyu Li
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, People’s Republic of China
- Ocean College, Beibu Gulf University, Qinzhou, Guangxi, People’s Republic of China
| |
Collapse
|
3
|
Arndts K, Wiszniewsky A, Neumann AL, Wiszniewsky K, Katawa G, Hoerauf A, Layland-Heni LE, Ritter M, Hübner MP. Differences of in vitro immune responses between patent and pre-patent Litomosoides sigmodontis-infected mice are independent of the filarial antigenic stimulus used. Parasitol Res 2024; 123:358. [PMID: 39436444 PMCID: PMC11496330 DOI: 10.1007/s00436-024-08365-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/03/2024] [Indexed: 10/23/2024]
Abstract
Lymphatic filariasis and onchocerciasis are neglected tropical diseases and cause significant public health problems in endemic countries, especially in sub-Saharan Africa. Since the human parasites are not viable in immune-competent mice, animal models have been developed, among them Litomosoides sigmodontis which permits a complete life cycle in BALB/c mice, including the development of patent infections with circulating microfilariae (Mf, the worm's offspring). To investigate the immunomodulatory properties of helminths in vitro, antigenic extracts can be prepared from different life cycle stages of the L. sigmodontis model, including adult worms, but the methods to prepare these antigens differ between research groups. This study analyzed whether different centrifugation methods during the preparation of an antigenic extract, the gender of used worms, or the different fractions (soluble or pellet) altered filarial-specific CD4+ T cell responses. These cells were isolated from pre-patent or patent/chronic infected mice, hence those without and those with Mf, respectively. Ex vivo immune responses were compared at these two different time points of the infection as well as the parasitic parameters. Worm burden and cell infiltration were elevated in the thoracic cavity (TC) and draining mediastinal lymph nodes at the pre-patent stage. Within the TC, eosinophils were significantly up-regulated at the earlier time point of infection which was further reflected by the eosinophil-related eotaxin-1 levels. Regarding the production of cytokines by re-stimulated CD4+ T cells in the presence of different antigen preparations, cytokine levels were comparable for all used extracts. Our data show that immune responses differ between pre-patent and patent filarial infection, but not in response to the different antigenic extracts themselves.
Collapse
Affiliation(s)
- Kathrin Arndts
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn (UKB), Venusberg-Campus 1, 53127, Bonn, Germany.
- German-West African Centre for Global Health and Pandemic Prevention (G-WAC), Partner Site Bonn, Bonn, Germany.
| | - Anna Wiszniewsky
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn (UKB), Venusberg-Campus 1, 53127, Bonn, Germany
| | - Anna-Lena Neumann
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn (UKB), Venusberg-Campus 1, 53127, Bonn, Germany
| | - Katharina Wiszniewsky
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn (UKB), Venusberg-Campus 1, 53127, Bonn, Germany
| | - Gnatoulma Katawa
- Unité de Recherche en Immunologie Et Immunomodulation (UR2IM), Université de Lomé, Ecole Supérieure Des Techniques Biologiques Et Alimentaires (ESTBA), Lomé, Togo
| | - Achim Hoerauf
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn (UKB), Venusberg-Campus 1, 53127, Bonn, Germany
- German-West African Centre for Global Health and Pandemic Prevention (G-WAC), Partner Site Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| | - Laura E Layland-Heni
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn (UKB), Venusberg-Campus 1, 53127, Bonn, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| | - Manuel Ritter
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn (UKB), Venusberg-Campus 1, 53127, Bonn, Germany
- German-West African Centre for Global Health and Pandemic Prevention (G-WAC), Partner Site Bonn, Bonn, Germany
| | - Marc P Hübner
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn (UKB), Venusberg-Campus 1, 53127, Bonn, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| |
Collapse
|
4
|
Rajamanickam A, Babu S. Helminth Infections and Diabetes: Mechanisms Accounting for Risk Amelioration. Annu Rev Nutr 2024; 44:339-355. [PMID: 38724017 DOI: 10.1146/annurev-nutr-061121-100742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2024]
Abstract
The global prevalence of type 2 diabetes mellitus (T2D) is increasing rapidly, with an anticipated 600 million cases by 2035. While infectious diseases such as helminth infections have decreased due to improved sanitation and health care, recent research suggests a link between helminth infections and T2D, with helminths such as Schistosoma, Nippostrongylus, Strongyloides, and Heligmosomoides potentially mitigating or slowing down T2D progression in human and animal models. Helminth infections enhance host immunity by promoting interactions between innate and adaptive immune systems. In T2D, type 1 immune responses are suppressed and type 2 responses are augmented, expanding regulatory T cells and innate immune cells, particularly type 2 immune cells and macrophages. This article reviews recent research shedding light on the favorable effects of helminth infections on T2D. The potential defense mechanisms identified include heightened insulin sensitivity and reduced inflammation. The synthesis of findings from studies investigating parasitic helminths and their derivatives underscores promising avenues for defense against T2D.
Collapse
Affiliation(s)
- Anuradha Rajamanickam
- National Institutes of Health-National Institute of Allergy and Infectious Diseases International Center for Excellence in Research, Chennai, India;
| | - Subash Babu
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
- National Institutes of Health-National Institute of Allergy and Infectious Diseases International Center for Excellence in Research, Chennai, India;
| |
Collapse
|
5
|
Karunakaran I, Surendar J, Ransmann P, Brühl M, Kowalski S, Frische V, Hmida J, Nachtsheim S, Hoerauf A, Wirtz DC, Hübner MP, Strauss AC, Schildberg FA. Seroprevalence and Association of Toxoplasma gondii with Bone Health in a Cohort of Osteopenia and Osteoporosis Patients. Biomedicines 2024; 12:1400. [PMID: 39061974 PMCID: PMC11274202 DOI: 10.3390/biomedicines12071400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/04/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
Considering the fact that Toxoplasma is a common parasite of humans and Toxoplasma bradyzoites can reside in skeletal muscle, T. gondii-mediated immune responses may modulate the progression and pathophysiology of another musculoskeletal disorder, osteoporosis. In the current study, we investigated the association of bone health and Toxoplasma gondii infection status. A total of 138 patients living in Germany with either osteopenia or osteoporosis were included in the study, and they were categorized into two groups, T. gondii uninfected (n = 74) and infected (n = 64), based on the presence of T. gondii-specific IgG antibodies. The demographic and clinical details of the study subjects were collected from the medical records. Logistic regression analysis was performed to delineate the association of bone health parameters with the infection status. The prevalence of toxoplasmosis was 46.4% in the study participants. The infected individuals with osteopenia and osteoporosis showed higher levels of mean spine and femoral T score, Z score, and bone mineral density (BMD), indicating improved bone health compared to the uninfected group. Logistic regression analysis showed that subjects with T. gondii infection displayed increased odds of having a higher mean femur T score, femur BMD, and femur Z score even after adjusting for age, creatinine, and urea levels. However, when the duration of drug intake for osteoporosis was taken into account, the association lost statistical significance. In summary, in this study, an improvement in osteopenia and osteoporosis was observed in Toxoplasma-infected patients, which may be partly due to the longer duration of drug intake for osteoporosis in the infected patient group.
Collapse
Affiliation(s)
- Indulekha Karunakaran
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, 53127 Bonn, Germany
| | - Jayagopi Surendar
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, 53127 Bonn, Germany
| | - Pia Ransmann
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, 53127 Bonn, Germany
| | - Marius Brühl
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, 53127 Bonn, Germany
| | - Silvia Kowalski
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, 53127 Bonn, Germany
| | - Victoria Frische
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, 53127 Bonn, Germany
| | - Jamil Hmida
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, 53127 Bonn, Germany
| | - Sabine Nachtsheim
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, 53127 Bonn, Germany
| | - Achim Hoerauf
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, 53127 Bonn, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, 53127 Bonn, Germany
| | - Dieter C. Wirtz
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, 53127 Bonn, Germany
| | - Marc P. Hübner
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, 53127 Bonn, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, 53127 Bonn, Germany
| | - Andreas C. Strauss
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, 53127 Bonn, Germany
| | - Frank A. Schildberg
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, 53127 Bonn, Germany
| |
Collapse
|
6
|
Sikder S, Pierce D, Sarkar ER, McHugh C, Quinlan KGR, Giacomin P, Loukas A. Regulation of host metabolic health by parasitic helminths. Trends Parasitol 2024; 40:386-400. [PMID: 38609741 DOI: 10.1016/j.pt.2024.03.006] [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: 02/12/2024] [Revised: 03/14/2024] [Accepted: 03/14/2024] [Indexed: 04/14/2024]
Abstract
Obesity is a worldwide pandemic and major risk factor for the development of metabolic syndrome (MetS) and type 2 diabetes (T2D). T2D requires lifelong medical support to limit complications and is defined by impaired glucose tolerance, insulin resistance (IR), and chronic low-level systemic inflammation initiating from adipose tissue. The current preventative strategies include a healthy diet, controlled physical activity, and medication targeting hyperglycemia, with underexplored underlying inflammation. Studies suggest a protective role for helminth infection in the prevention of T2D. The mechanisms may involve induction of modified type 2 and regulatory immune responses that suppress inflammation and promote insulin sensitivity. In this review, the roles of helminths in counteracting MetS, and prospects for harnessing these protective mechanisms for the development of novel anti-diabetes drugs are discussed.
Collapse
Affiliation(s)
- Suchandan Sikder
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland 4878, Australia.
| | - Doris Pierce
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland 4878, Australia
| | - Eti R Sarkar
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland 4878, Australia; College of Public Health, Medical and Veterinary Sciences, James Cook University, Cairns, Queensland 4878, Australia
| | - Connor McHugh
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland 4878, Australia; College of Public Health, Medical and Veterinary Sciences, James Cook University, Cairns, Queensland 4878, Australia
| | - Kate G R Quinlan
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, 2052, Australia
| | - Paul Giacomin
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland 4878, Australia; Macrobiome Therapeutics Pty Ltd, Cairns, Queensland 4878, Australia
| | - Alex Loukas
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland 4878, Australia; Macrobiome Therapeutics Pty Ltd, Cairns, Queensland 4878, Australia
| |
Collapse
|
7
|
Singh I, Hoti SL, Chauhan N, Joshi RK, Prasad TSK, Sarikhani M, Kaushik M, Unger BS, Jadhav P, Modi PK. Immunomodulation of streptozotocin induced Type 1 diabetes mellitus in mouse model by Macrophage migration inhibitory factor-2 (MIF-2) homologue of human lymphatic filarial parasite, Wuchereria bancrofti. Acta Trop 2024; 252:107142. [PMID: 38331083 DOI: 10.1016/j.actatropica.2024.107142] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 02/05/2024] [Accepted: 02/05/2024] [Indexed: 02/10/2024]
Abstract
Helminth parasites modulate the host immune system to ensure a long-lasting asymptomatic form of infection generally, mediated by the secretion of immunomodulatory molecules and one such molecule is a homologue of human host cytokine, Macrophage migratory Inhibitory Factor (hMIF). In this study, we sought to understand the role of homologue of hMIF from the lymphatic filarial parasite, Wuchereria bancrofti (Wba-MIF2), in the immunomodulation of the Streptozotocin (STZ)-induced Type1 Diabetes Mellitus (T1DM) animal model. Full-length recombinant Wba-MIF2 was expressed and found to have both oxidoreductase and tautomerase activities. Wba-MIF2 recombinant protein was treated to STZ induced T1DM animals, and after 5 weeks pro-inflammatory (IL-1, IL-2, IL-6, TNF-α, IFN-γ) and anti-inflammatory (IL-4, IL-10) cytokines and gene expressions were determined in sera samples and spleen respectively. Pro-inflammatory and anti-inflammatory cytokine levels were significantly (p<0.05) up-regulated and down-regulated respectively, in the STZ-T1DM animals, as compared to treated groups. Histopathology showed macrophage infiltration and greater damage of islets of beta cells in the pancreatic tissue of STZ-T1DM animals, than Wba-MIF2 treated STZ-T1DM animals. The present study clearly showed the potential of Wba-MIF2 as an immunomodulatory molecule, which could modulate the host immune system in the STZ-T1DM mice model from a pro-inflammatory to anti-inflammatory milieu.
Collapse
Affiliation(s)
- Ishwar Singh
- ICMR-National Institute of Traditional Medicine, Belagavi 590010, India Karnataka, India; KLE Academy of Higher Education and Research, Belagavi 590010, India Karnataka, India
| | - S L Hoti
- ICMR-National Institute of Traditional Medicine, Belagavi 590010, India Karnataka, India.
| | - Nikhil Chauhan
- ICMR-National Institute of Traditional Medicine, Belagavi 590010, India Karnataka, India
| | - R K Joshi
- ICMR-National Institute of Traditional Medicine, Belagavi 590010, India Karnataka, India
| | - T S Keshava Prasad
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore 575007, Karnataka, India
| | | | - Meenakshi Kaushik
- ICMR-National Institute of Traditional Medicine, Belagavi 590010, India Karnataka, India
| | - Banappa S Unger
- ICMR-National Institute of Traditional Medicine, Belagavi 590010, India Karnataka, India
| | - Pankaj Jadhav
- Indian Institute of Science, Bangalore 560012, Karnataka, India
| | - Prashant Kumar Modi
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore 575007, Karnataka, India
| |
Collapse
|
8
|
Tong M, Yang X, Qiao Y, Liu G, Ge H, Huang G, Wang Y, Yang Y, Fan W. Serine protease inhibitor from the muscle larval Trichinella spiralis ameliorates non-alcoholic fatty liver disease in mice via anti-inflammatory properties and gut-liver crosstalk. Biomed Pharmacother 2024; 172:116223. [PMID: 38325266 DOI: 10.1016/j.biopha.2024.116223] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/22/2024] [Accepted: 01/29/2024] [Indexed: 02/09/2024] Open
Abstract
Trichinella spiralis is recognized for its ability to regulate host immune responses. The serine protease inhibitor of T. spiralis (Ts-SPI) participates in T. spiralis-mediated immunoregulatory effects. Studies have shown that helminth therapy exhibits therapeutic effects on metabolic diseases. In addition, we previously found that T. spiralis-derived crude antigens could alleviate diet-induced obesity. Thus, Ts-SPI was hypothesized to alleviate non-alcoholic fatty liver disease (NAFLD). Herein, recombinant Ts-SPI (rTs-SPI) was prepared from the muscle larvae T. spiralis. The relative molecular mass of rTs-SPI was approximately 35,000 Da, and western blot analysis indicated good immunoreactivity. rTs-SPI ameliorated hepatic steatosis, inflammation, and pyroptosis in NAFLD mice, which validated the hypothesis. rTs-SPI also reduced macrophage infiltration, significantly expanded Foxp3+ Treg population, and inactivated TLR4/NF-κB/NLRP3 signaling in the liver. Furthermore, rTs-SPI treatment significantly shifted the gut microbiome structure, with a remarkable increase in beneficial bacteria and reduction in harmful bacteria to improve gut barrier integrity. Finally, Abx-treated mice and FMT confirmed that gut-liver crosstalk contributed to NAFLD improvement after rTs-SPI treatment. Taken together, Taken together, these findings suggest that rTs-SPI exerts therapeutic effects in NAFLD via anti-inflammatory activity and gut-liver crosstalk.
Collapse
Affiliation(s)
- Mingwei Tong
- School of Basic Medical Sciences, Shanxi Medical University, Jinzhong 030619, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, and Shanxi Key Laboratory of Cellular Physiology, Taiyuan 030001, China.
| | - Xiaodan Yang
- School of Basic Medical Sciences, Shanxi Medical University, Jinzhong 030619, China
| | - Yuyu Qiao
- School of Basic Medical Sciences, Shanxi Medical University, Jinzhong 030619, China
| | - Ge Liu
- School of Basic Medical Sciences, Shanxi Medical University, Jinzhong 030619, China
| | - Huihui Ge
- School of Basic Medical Sciences, Shanxi Medical University, Jinzhong 030619, China
| | - Guangrong Huang
- School of Basic Medical Sciences, Shanxi Medical University, Jinzhong 030619, China
| | - Yanhong Wang
- School of Basic Medical Sciences, Shanxi Medical University, Jinzhong 030619, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, and Shanxi Key Laboratory of Cellular Physiology, Taiyuan 030001, China
| | - Yong Yang
- School of Basic Medical Sciences, Shanxi Medical University, Jinzhong 030619, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, and Shanxi Key Laboratory of Cellular Physiology, Taiyuan 030001, China.
| | - Weiping Fan
- School of Basic Medical Sciences, Shanxi Medical University, Jinzhong 030619, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, and Shanxi Key Laboratory of Cellular Physiology, Taiyuan 030001, China.
| |
Collapse
|
9
|
Camaya I, O’Brien B, Donnelly S. How do parasitic worms prevent diabetes? An exploration of their influence on macrophage and β-cell crosstalk. Front Endocrinol (Lausanne) 2023; 14:1205219. [PMID: 37564976 PMCID: PMC10411736 DOI: 10.3389/fendo.2023.1205219] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 07/10/2023] [Indexed: 08/12/2023] Open
Abstract
Diabetes is the fastest growing chronic disease globally, with prevalence increasing at a faster rate than heart disease and cancer. While the disease presents clinically as chronic hyperglycaemia, two distinct subtypes have been recognised. Type 1 diabetes (T1D) is characterised as an autoimmune disease in which the insulin-producing pancreatic β-cells are destroyed, and type 2 diabetes (T2D) arises due to metabolic insufficiency, in which inadequate amounts of insulin are produced, and/or the actions of insulin are diminished. It is now apparent that pro-inflammatory responses cause a loss of functional β-cell mass, and this is the common underlying mechanism of both T1D and T2D. Macrophages are the central immune cells in the pathogenesis of both diseases and play a major role in the initiation and perpetuation of the proinflammatory responses that compromise β-cell function. Furthermore, it is the crosstalk between macrophages and β-cells that orchestrates the inflammatory response and ensuing β-cell dysfunction/destruction. Conversely, this crosstalk can induce immune tolerance and preservation of β-cell mass and function. Thus, specifically targeting the intercellular communication between macrophages and β-cells offers a unique strategy to prevent/halt the islet inflammatory events underpinning T1D and T2D. Due to their potent ability to regulate mammalian immune responses, parasitic worms (helminths), and their excretory/secretory products, have been examined for their potential as therapeutic agents for both T1D and T2D. This research has yielded positive results in disease prevention, both clinically and in animal models. However, the focus of research has been on the modulation of immune cells and their effectors. This approach has ignored the direct effects of helminths and their products on β-cells, and the modulation of signal exchange between macrophages and β-cells. This review explores how the alterations to macrophages induced by helminths, and their products, influence the crosstalk with β-cells to promote their function and survival. In addition, the evidence that parasite-derived products interact directly with endocrine cells to influence their communication with macrophages to prevent β-cell death and enhance function is discussed. This new paradigm of two-way metabolic conversations between endocrine cells and macrophages opens new avenues for the treatment of immune-mediated metabolic disease.
Collapse
Affiliation(s)
| | | | - Sheila Donnelly
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| |
Collapse
|
10
|
Lenz B, Andrew BA, Ritter M, Karunakaran I, Gandjui NVT, Nchang LC, Surendar J, Ebob AOB, Ehrens A, Klarmann-Schulz U, Ricchiuto A, Kuehlwein JM, Fombad FF, Ngwa AM, Katcho TD, Hoerauf A, Wanji S, Hübner MP. The design and development of a study protocol to investigate Onchocerca volvulus, Loa loa and Mansonella perstans-mediated modulation of the metabolic and immunological profile in lean and obese individuals in Cameroon. PLoS One 2023; 18:e0285689. [PMID: 37267236 DOI: 10.1371/journal.pone.0285689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 04/10/2023] [Indexed: 06/04/2023] Open
Abstract
BACKGROUND Life-style metabolic diseases are steadily rising, not only in developed countries, but also in low- and middle-income countries, presenting a global health problem. Metabolic disorders like type 2 diabetes and cardiovascular diseases are among the ten leading causes of death defined by the WHO in 2019. Results from animal and observational human studies suggest a connection between the decline in human helminth infections and rise of life-style-associated metabolic diseases in developing regions. This trial was designed to investigate filarial infections and their impact on metabolic diseases in Cameroon. We hypothesize that the induction of regulatory immune responses during filarial infection reduces obesity-induced low-grade inflammatory immune responses and thereby improves metabolic parameters, whereas anthelmintic treatment abolishes this protective effect. METHODS/DESIGN Participants infected with Mansonella perstans, Onchocerca volvulus and/or Loa loa being lean (BMI <25), overweight (BMI >25 and <30) or clinically obese (BMI ≥30) from Littoral regions of Cameroon will be evaluated for their parasitological, immunological, metabolic and biochemical profile before and after treatment of their parasitic infections. Anthropomorphic measurements and a detailed questionnaire will complement our analysis. The investigation will assess blood immune cell populations, serum adipokines and cytokines that could be influenced by the parasite infection and/or metabolic diseases. Further, parameters like blood glucose, homeostatic model assessment of insulin resistance (HOMA-IR), circulating lipids and circulating makers of liver function will be monitored. Parameters will be assessed before treatment, 12 and 18 months after treatment. CONCLUSION The focus of this study is to obtain a comprehensive metabolic profile of the participants in rural areas of Cameroon and to investigate the relationship between filarial immunomodulation and metabolic diseases. This study will elucidate the effect of anti-filarial treatment on the metabolic and immunological parameters that partake in the development of insulin resistance, narrowing in on a potential protective effect of filarial infections on metabolic diseases. TRIAL REGISTRATION doi.org/10.1186/ISRCTN43845142, ISRCTN43845142 February 2020 Trial title Effects of filarial parasite infection on type 2 diabetes Issue date: 27.10.22, V.1.
Collapse
Affiliation(s)
- Benjamin Lenz
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Beng Amuam Andrew
- Parasite and Vector Biology Research Unit, Department of Microbiology and Parasitology, Faculty of Science, University of Buea, Buea, Cameroon
- Research Foundation in Tropical Diseases and the Environment, Buea, Cameroon
| | - Manuel Ritter
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Indulekha Karunakaran
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Narcisse Victor Tchamatchoua Gandjui
- Parasite and Vector Biology Research Unit, Department of Microbiology and Parasitology, Faculty of Science, University of Buea, Buea, Cameroon
- Research Foundation in Tropical Diseases and the Environment, Buea, Cameroon
| | - Lucy Cho Nchang
- Parasite and Vector Biology Research Unit, Department of Microbiology and Parasitology, Faculty of Science, University of Buea, Buea, Cameroon
- Research Foundation in Tropical Diseases and the Environment, Buea, Cameroon
| | - Jayagopi Surendar
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Anita Obi Bate Ebob
- Parasite and Vector Biology Research Unit, Department of Microbiology and Parasitology, Faculty of Science, University of Buea, Buea, Cameroon
- Research Foundation in Tropical Diseases and the Environment, Buea, Cameroon
| | - Alexandra Ehrens
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), Bonn, Germany
| | - Ute Klarmann-Schulz
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), Bonn, Germany
| | - Arcangelo Ricchiuto
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Janina M Kuehlwein
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), Bonn, Germany
| | - Fanny Fri Fombad
- Parasite and Vector Biology Research Unit, Department of Microbiology and Parasitology, Faculty of Science, University of Buea, Buea, Cameroon
- Research Foundation in Tropical Diseases and the Environment, Buea, Cameroon
| | - Ambe Marius Ngwa
- Parasite and Vector Biology Research Unit, Department of Microbiology and Parasitology, Faculty of Science, University of Buea, Buea, Cameroon
- Research Foundation in Tropical Diseases and the Environment, Buea, Cameroon
| | - Tatiana Djikeussi Katcho
- Parasite and Vector Biology Research Unit, Department of Microbiology and Parasitology, Faculty of Science, University of Buea, Buea, Cameroon
- Research Foundation in Tropical Diseases and the Environment, Buea, Cameroon
| | - Achim Hoerauf
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), Bonn, Germany
- German-West African Centre for Global Health and Pandemic Prevention (G-WAC), Bonn, Germany
| | - Samuel Wanji
- Parasite and Vector Biology Research Unit, Department of Microbiology and Parasitology, Faculty of Science, University of Buea, Buea, Cameroon
- Research Foundation in Tropical Diseases and the Environment, Buea, Cameroon
| | - Marc P Hübner
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), Bonn, Germany
| |
Collapse
|
11
|
Myhill LJ, Williams AR. Diet-microbiota crosstalk and immunity to helminth infection. Parasite Immunol 2023; 45:e12965. [PMID: 36571323 DOI: 10.1111/pim.12965] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 12/06/2022] [Accepted: 12/10/2022] [Indexed: 12/27/2022]
Abstract
Helminths are large multicellular parasites responsible for widespread chronic disease in humans and animals. Intestinal helminths live in close proximity with the host gut microbiota and mucosal immune network, resulting in reciprocal interactions that closely influence the course of infections. Diet composition may strongly regulate gut microbiota composition and intestinal immune function and therefore may play a key role in modulating anti-helminth immune responses. Characterizing the multitude of interactions that exist between different dietary components (e.g., dietary fibres), immune cells, and the microbiota, may shed new light on regulation of helminth-specific immunity. This review focuses on the current knowledge of how metabolism of dietary components shapes immune response during helminth infection, and how this information may be potentially harnessed to design new therapeutics to manage parasitic infections and associated diseases.
Collapse
Affiliation(s)
- Laura J Myhill
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Andrew R Williams
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
12
|
Karunakaran I, Ritter M, Pfarr K, Klarmann-Schulz U, Debrah AY, Debrah LB, Katawa G, Wanji S, Specht S, Adjobimey T, Hübner MP, Hoerauf A. Filariasis research - from basic research to drug development and novel diagnostics, over a decade of research at the Institute for Medical Microbiology, Immunology and Parasitology, Bonn, Germany. FRONTIERS IN TROPICAL DISEASES 2023; 4:1126173. [PMID: 38655130 PMCID: PMC7615856 DOI: 10.3389/fitd.2023.1126173] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024] Open
Abstract
Filariae are vector borne parasitic nematodes, endemic in tropical and subtropical regions causing avoidable infections ranging from asymptomatic to stigmatizing and disfiguring disease. The filarial species that are the major focus of our institution's research are Onchocerca volvulus causing onchocerciasis (river blindness), Wuchereria bancrofti and Brugia spp. causing lymphatic filariasis (elephantiasis), Loa loa causing loiasis (African eye worm), and Mansonella spp causing mansonellosis. This paper aims to showcase the contribution of our institution and our collaborating partners to filarial research and covers decades of long research spanning basic research using the Litomosoides sigmodontis animal model to development of drugs and novel diagnostics. Research with the L. sigmodontis model has been extensively useful in elucidating protective immune responses against filariae as well as in identifying the mechanisms of filarial immunomodulation during metabolic, autoimmune and infectious diseases. The institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn (UKB), Bonn, Germany has also been actively involved in translational research in contributing to the identification of new drug targets and pre-clinical drug research with successful and ongoing partnership with sub-Saharan Africa, mainly Ghana (the Kumasi Centre for Collaborative Research (KCCR)), Cameroon (University of Buea (UB)) and Togo (Laboratoire de Microbiologie et de Contrôle de Qualité des Denrées Alimentaires (LAMICODA)), Asia and industry partners. Further, in the direction of developing novel diagnostics that are sensitive, time, and labour saving, we have developed sensitive qPCRs as well as LAMP assays and are currently working on artificial intelligence based histology analysis for onchocerciasis. The article also highlights our ongoing research and the need for novel animal models and new drug targets.
Collapse
Affiliation(s)
- Indulekha Karunakaran
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn (UKB), Bonn, Germany
| | - Manuel Ritter
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn (UKB), Bonn, Germany
| | - Kenneth Pfarr
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn (UKB), Bonn, Germany
- German Center for Infection Research (DZIF), partner site Bonn-Cologne, Bonn, Germany
| | - Ute Klarmann-Schulz
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn (UKB), Bonn, Germany
- German Center for Infection Research (DZIF), partner site Bonn-Cologne, Bonn, Germany
| | - Alexander Yaw Debrah
- Faculty of Allied Health Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Kumasi Center for Collaborative Research (KCCR), Kumasi, Ghana
| | - Linda Batsa Debrah
- Kumasi Center for Collaborative Research (KCCR), Kumasi, Ghana
- Department of Clinical Microbiology, School of Medicine and Dentistry, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Gnatoulma Katawa
- Unité de Recherche en Immunologie et Immunomodulation (UR2IM)/Laboratoire de Microbiologie et de Contrôle de Qualité des Denrées Alimentaires (LAMICODA), Ecole Supérieure des Techniques Biologiques et Alimentaires, Université de Lomé, Lomé, Togo
| | - Samuel Wanji
- Parasites and Vector Research Unit (PAVRU), Department of Microbiology and Parasitology, University of Buea, Buea, Cameroon
- Research Foundation in Tropical Diseases and Environment (REFOTDE), Buea, Cameroon
| | - Sabine Specht
- Drugs for Neglected Diseases initiative, Geneva, Switzerland
| | - Tomabu Adjobimey
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn (UKB), Bonn, Germany
| | - Marc P Hübner
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn (UKB), Bonn, Germany
- German Center for Infection Research (DZIF), partner site Bonn-Cologne, Bonn, Germany
| | - Achim Hoerauf
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn (UKB), Bonn, Germany
- German Center for Infection Research (DZIF), partner site Bonn-Cologne, Bonn, Germany
| |
Collapse
|
13
|
Liu X, Jiang Y, Ye J, Wang X. Helminth infection and helminth-derived products: A novel therapeutic option for non-alcoholic fatty liver disease. Front Immunol 2022; 13:999412. [PMID: 36263053 PMCID: PMC9573989 DOI: 10.3389/fimmu.2022.999412] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/15/2022] [Indexed: 11/13/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is closely related to obesity, diabetes, and metabolic syndrome (MetS), and it has become the most common chronic liver disease. Helminths have co-evolved with humans, inducing multiple immunomodulatory mechanisms to modulate the host's immune system. By using their immunomodulatory ability, helminths and their products exhibit protection against various autoimmune and inflammatory diseases, including obesity, diabetes, and MetS, which are closely associated with NAFLD. Here, we review the pathogenesis of NAFLD from abnormal glycolipid metabolism, inflammation, and gut dysbiosis. Correspondingly, helminths and their products can treat or relieve these NAFLD-related diseases, including obesity, diabetes, and MetS, by promoting glycolipid metabolism homeostasis, regulating inflammation, and restoring the balance of gut microbiota. Considering that a large number of clinical trials have been carried out on helminths and their products for the treatment of inflammatory diseases with promising results, the treatment of NAFLD and obesity-related diseases by helminths is also a novel direction and strategy.
Collapse
Affiliation(s)
- Xi Liu
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yuyun Jiang
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Jixian Ye
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xuefeng Wang
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Department of Nuclear Medicine and Institute of Digestive Diseases, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
14
|
Molecular Mechanism of YuPingFeng in the Treatment of Asthma Based on Network Pharmacology and Molecular Docking Technology. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:7364126. [PMID: 36105239 PMCID: PMC9467798 DOI: 10.1155/2022/7364126] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 07/26/2022] [Indexed: 11/17/2022]
Abstract
Objective To explore the molecular targets and mechanism of YuPingFeng (YPF) for the treatment of asthma by using network pharmacology and molecular docking. Methods The potential active ingredients and relevant targets of YPF were obtained from the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP). Asthma-related gene targets were retrieved from GeneCards, OMIM, DrugBank, PharmGKB, and TTD databases. The protein-protein (PPI) network between YPF and asthma common targets was constructed by SRING online database and Cytoscape software. GO and KEGG analyses were performed to explore the complicated molecular biological processes and potential pathways. Finally, a molecular docking approach was carried out to verify the results. Results We obtained 100 potential targets of the 35 active ingredients in YPF and 1610 asthma-related targets. 60 YPF-asthma common targets were selected to perform PPI analysis. Seven core genes were screened based on two topological calculation methods. GO and KEGG results showed that the main pathways of YPF in treating asthma include TNF signaling pathway and PI3K-Akt signaling pathway. Finally, the molecular docking results indicated that the key ingredients of YPF had a good affinity with the relevant core genes. Conclusion This study reflects the multicomponent, multitarget, and multipathway characteristics of YPF in treating asthma, providing a theoretical and scientific basis for the intervention of asthma by traditional Chinese medicine YPF.
Collapse
|
15
|
Cytokines, Chemokines, Insulin and Haematological Indices in Type 2 Diabetic Male Sprague Dawley Rats Infected with Trichinella zimbabwensis. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12157743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Diabetes mellitus is a chronic metabolic disease induced by the inability to control high blood glucose level. Helminth-induced immunomodulation has been reported to prevent or delay the onset of type 2 diabetes mellitus (T2DM), which, in turn, ameliorates insulin sensitivity. Therefore, there is a need to understand the underlying mechanisms utilized by helminths in metabolism and the induction of immuno-inflammatory responses during helminthic infection and T2DM comorbidity. This study aimed at using a laboratory animal model to determine the cytokines, chemokines and haematological indices in diabetic (T2DM) male Sprague Dawley (SD) rats infected with Trichinella zimbabwensis. One hundred and two male SD rats (160–180 g) were randomly selected into three experimental groups (i. T2DM-induced group (D) ii. T. zimbabwensis infected + T2DM group (TzD) and iii. T. zimbabwensis-infected group (Tz)). Rats selected for the D group and TzD group were injected with 40 mg/kg live weight of streptozotocin (STZ) intraperitoneally to induce T2DM, while animals in the Tz and TzD group were infected with T. zimbabwensis. Results showed that adult T. zimbabwensis worm loads and mean T. zimbabwensis larvae per gram (lpg) of rat muscle were significantly higher (p < 0.001) in the Tz group when compared to the TzD group. Blood glucose levels in the D group were significantly higher (p < 0.001) compared to the TzD group. An increase in insulin concentration was observed among the TzD group when compared to the D group. Liver and muscle glycogen decreased in the D when compared to the TzD group. A significant increase (p < 0.05) in red blood cells (RBCs) was observed in the D group when compared to the TzD and Tz groups. An increase in haematocrit, haemoglobin, white blood cells (WBCs), platelet, neutrophils and monocyte were observed in the D group when compared to the TzD group. TNF-α, IFN-γ, IL-4, IL-10 and IL-13 concentrations were elevated in the TzD group when compared to the D and Tz groups, while IL-6 concentration showed a significant reduction in the Tz when compared to the D and the TzD groups. A significant increase in CCL5 in the D and TzD groups was observed in comparison to the Tz group. CXCL10 and CCL11 concentration also showed an increase in the TzD group in comparison to the Tz and the D groups. Overall, our results confirm that T. zimbabwensis, a parasite which produces tissue-dwelling larvae in the host, regulates T2DM driven inflammation to mediate a positive protective effect against T2DM outcomes.
Collapse
|
16
|
Queiroz-Glauss CP, Vieira MS, Gonçalves-Pereira MH, Almeida SS, Freire RH, Gomes MA, Alvarez-Leite JI, Santiago HC. Helminth infection modulates number and function of adipose tissue Tregs in high fat diet-induced obesity. PLoS Negl Trop Dis 2022; 16:e0010105. [PMID: 35499991 PMCID: PMC9098094 DOI: 10.1371/journal.pntd.0010105] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 05/12/2022] [Accepted: 04/01/2022] [Indexed: 11/23/2022] Open
Abstract
Background Epidemiological and experimental studies have shown a protective effect of helminth infections in weight gain and against the development of metabolic dysfunctions in the host. However, the mechanisms Treg cells exert in the helminth-obesity interface has been poorly investigated. The present study aimed to verify the influence of Heligmosomoides polygyrus infection in early stages of high fat diet-induced obesity. Principal findings The presence of infection was able to prevent exacerbated weight gain in mice fed with high fat diet when compared to non-infected controls. In addition, infected animals displayed improved insulin sensitivity and decreased fat accumulation in the liver. Obesity-associated inflammation was reduced in the presence of infection, demonstrated by lower levels of leptin and resistin, lower infiltration of Th1 and Th17 cells in adipose tissue, higher expression of IL10 and adiponectin, increased infiltration of Th2 and eosinophils in adipose tissue of infected animals. Of note, the parasite infection was associated with increased Treg frequency in adipose tissue which showed higher expression of cell surface markers of function and activation, like LAP and CD134. The infection could also increase adipose Treg suppressor function in animals on high fat diet. Conclusion These data suggest that H. polygyrus modulates adipose tissue Treg cells with implication for weight gain and metabolic syndrome. Helminth infections are known to modulate the immune system being responsible for protecting the host from developing allergic and autoimmune disorders (Hygiene Hypothesis). We hypothesized that the same immunomodulatory effect could have an impact on immunometabolic diseases, such as obesity and its linked diseases such as type 2 diabetes. Weight disorders have reached epidemic levels, nearly tripling since 1975 and being responsible for almost 5 million premature deaths each year, but have been spared in areas of high helminth prevalence. To test our hypothesis C57BL/6 male mice were fed control or high fat diet, for five weeks, in the presence or not of infection with the worm Heligmosomoides polygyrus. Weight gain, development of metabolic disorders, inflammation and cellular migration to the adipose tissue were evaluated. In accordance with our hypothesis, we found that the presence of infection prevented the exacerbated weight gain and also improved metabolic parameters in animals fed a high fat diet. This was associated with the infection’s ability to modulate parameters of a cell responsible for regulatory functions: Tregs. In the light of these findings, helminth infection could be protective against weight gain and metabolic disturbances.
Collapse
Affiliation(s)
- Camila P. Queiroz-Glauss
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mariana S. Vieira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Marcela Helena Gonçalves-Pereira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Stephanie S. Almeida
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Rachel H. Freire
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Maria A. Gomes
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Jacqueline I. Alvarez-Leite
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Helton C. Santiago
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- * E-mail:
| |
Collapse
|
17
|
Administration of Hookworm Excretory/Secretory Proteins Improves Glucose Tolerance in a Mouse Model of Type 2 Diabetes. Biomolecules 2022; 12:biom12050637. [PMID: 35625566 PMCID: PMC9138508 DOI: 10.3390/biom12050637] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/19/2022] [Accepted: 04/22/2022] [Indexed: 01/27/2023] Open
Abstract
Diabetes is recognised as the world’s fastest growing chronic condition globally. Helminth infections have been shown to be associated with a lower prevalence of type 2 diabetes (T2D), in part due to their ability to induce a type 2 immune response. Therefore, to understand the molecular mechanisms that underlie the development of T2D-induced insulin resistance, we treated mice fed on normal or diabetes-promoting diets with excretory/secretory products (ES) from the gastrointestinal helminth Nippostrongylus brasiliensis. We demonstrated that treatment with crude ES products from adult worms (AES) or infective third-stage larvae (L3ES) from N. brasiliensis improved glucose tolerance and attenuated body weight gain in mice fed on a high glycaemic index diet. N. brasiliensis ES administration to mice was associated with a type 2 immune response measured by increased eosinophils and IL-5 in peripheral tissues but not IL-4, and with a decrease in the level of IL-6 in adipose tissue and corresponding increase in IL-6 levels in the liver. Moreover, treatment with AES or L3ES was associated with significant changes in the community composition of the gut microbiota at the phylum and order levels. These data highlight a role for N. brasiliensis ES in modulating the immune response associated with T2D, and suggest that N. brasiliensis ES contain molecules with therapeutic potential for treating metabolic syndrome and T2D.
Collapse
|
18
|
Dai M, Yang X, Yu Y, Pan W. Helminth and Host Crosstalk: New Insight Into Treatment of Obesity and Its Associated Metabolic Syndromes. Front Immunol 2022; 13:827486. [PMID: 35281054 PMCID: PMC8913526 DOI: 10.3389/fimmu.2022.827486] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/02/2022] [Indexed: 12/16/2022] Open
Abstract
Obesity and its associated Metabolic Syndromes (Mets) represent a global epidemic health problem. Metabolic inflammation, lipid accumulation and insulin resistance contribute to the progression of these diseases, thereby becoming targets for drug development. Epidemiological data have showed that the rate of helminth infection negatively correlates with the incidence of obesity and Mets. Correspondingly, numerous animal experiments and a few of clinic trials in human demonstrate that helminth infection or its derived molecules can mitigate obesity and Mets via induction of macrophage M2 polarization, inhibition of adipogenesis, promotion of fat browning, and improvement of glucose tolerance, insulin resistance and metabolic inflammation. Interestingly, sporadic studies also uncover that several helminth infections can reshape gut microbiota of hosts, which is intimately implicated in the pathogenesis of obesity and Mets. Overall, these findings indicate that the crosstalk between helminth and hosts may be a novel direction for obesity and Mets therapy. The present article reviews the molecular mechanism of how helminth masters immunity and metabolism in obesity.
Collapse
Affiliation(s)
- Mengyu Dai
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- The Second Clinical Medicine, Xuzhou Medical University, Xuzhou, China
- National Demonstration Center for Experimental Basic Medical Science Education (Xuzhou Medical University), Xuzhou, China
| | - Xiaoying Yang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Yinghua Yu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Wei Pan, ; Yinghua Yu,
| | - Wei Pan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Wei Pan, ; Yinghua Yu,
| |
Collapse
|
19
|
Novel Insights into the Immunomodulatory Effects of Caryophyllane Sesquiterpenes: A Systematic Review of Preclinical Studies. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12052292] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
Immunomodulation is a key factor in the homeostasis of organisms, both for physiological and inflammatory conditions. In this context, great attention has been devoted to immunomodulant agents, which can boost or modulate the immune system, thus favoring disease relief. The present systematic review is focused on the immunomodulatory properties of plant-based caryophyllane sesquiterpenes, which are unique natural compounds widely studied due to their multiple and pleiotropic bioactivities. Despite lacking clinical evidence, the selected studies highlighted the ability of these substances, especially β-caryophyllene and α-humulene, to modulate the immune system of both in vitro and in vivo models of disease, such as neurodegenerative and inflammatory-based diseases, cancer, and allergies; moreover, some mechanistic hypotheses have been made too. The present overview suggests a further interest in immunomodulation by caryophyllane sesquiterpenes as a possible novel strategy for immune-based diseases or as an adjuvant treatment and encourages further high-quality studies, using high-purity compounds, to better clarify the mechanisms accounting for these properties and to support a further pharmaceutical development.
Collapse
|
20
|
Sibi JM, Mohan V, Deepa M, Babu S, Aravindhan V. Modulatory effect of filarial infection on the systemic hormone levels in subjects with metabolic syndrome (DM-LF5). Front Endocrinol (Lausanne) 2022; 13:1011942. [PMID: 36482987 PMCID: PMC9723321 DOI: 10.3389/fendo.2022.1011942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 11/01/2022] [Indexed: 11/23/2022] Open
Abstract
AIM Metabolic syndrome (MS) refers to a group of co-morbidities which include central obesity, hypertension, hyperglycemia and dyslipidemia. Previously, we reported that childhood lymphatic filariasis (LF) confers significant protection against type-1 and type-2 forms of diabetes, by means of immunomodulation. In the present study, we studied the effect of LF on endocrine dysfunction in MS and Non-MS patients in baseline and after 10 years of follow-up. METHODS We quantified the serum levels of pancreatic hormones (insulin and glucagon), incretins (Ghrelin, GIP and GLP-1) and adipokines (leptin, adiponectin, adipsin, visfatin, PAI-1 and resistin) by multiplex bead array system. RESULTS MS (both LF- and LF+) subjects had increased insulin levels compared to NMS (both LF- and LF+) subjects. MS-LF+ subjects had significantly increased levels of glucagon, ghrelin, GIP and GLP-1 and decreased levels of adipsin, compared to MS-LF- subjects. Interestingly this effect was short-lived and was not seen in the follow-up samples. CONCLUSION Overall, LF infection might confer limited short-term beneficial effects against MS, by means of modulating the incretin levels,either directly or indirectly.
Collapse
Affiliation(s)
- Joy Manohar Sibi
- Department of Genetics, Dr. A. L. Mudaliar Post Graduate Institute of Basic Medical Sciences, University of Madras, Chennai, India
| | - Viswanathan Mohan
- Madras Diabetes Research Foundation and Dr. Mohan’s Diabetes Specialties Centre, ICMR Center for Advanced Research on Diabetes and IDF Centre of Excellence in Diabetes Care, Chennai, India
| | - Mohan Deepa
- Madras Diabetes Research Foundation and Dr. Mohan’s Diabetes Specialties Centre, ICMR Center for Advanced Research on Diabetes and IDF Centre of Excellence in Diabetes Care, Chennai, India
| | - Subash Babu
- National Institute of Health-International Centre for Excellence in Research, National Institute for Research in Tuberculosis, Chennai, India
| | - Vivekanandhan Aravindhan
- Department of Genetics, Dr. A. L. Mudaliar Post Graduate Institute of Basic Medical Sciences, University of Madras, Chennai, India
- *Correspondence: Vivekanandhan Aravindhan,
| |
Collapse
|
21
|
Risch F, Ritter M, Hoerauf A, Hübner MP. Human filariasis-contributions of the Litomosoides sigmodontis and Acanthocheilonema viteae animal model. Parasitol Res 2021; 120:4125-4143. [PMID: 33547508 PMCID: PMC8599372 DOI: 10.1007/s00436-020-07026-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/20/2020] [Indexed: 12/16/2022]
Abstract
Filariae are vector-borne parasitic nematodes that are endemic worldwide, in tropical and subtropical regions. Important human filariae spp. include Onchocerca volvulus, Wuchereria bancrofti and Brugia spp., and Loa loa and Mansonella spp. causing onchocerciasis (river blindness), lymphatic filariasis (lymphedema and hydrocele), loiasis (eye worm), and mansonelliasis, respectively. It is estimated that over 1 billion individuals live in endemic regions where filarial diseases are a public health concern contributing to significant disability adjusted life years (DALYs). Thus, efforts to control and eliminate filarial diseases were already launched by the WHO in the 1970s, especially against lymphatic filariasis and onchocerciasis, and are mainly based on mass drug administration (MDA) of microfilaricidal drugs (ivermectin, diethylcarbamazine, albendazole) to filarial endemic areas accompanied with vector control strategies with the goal to reduce the transmission. With the United Nations Sustainable Development Goals (SDGs), it was decided to eliminate transmission of onchocerciasis and stop lymphatic filariasis as a public health problem by 2030. It was also requested that novel drugs and treatment strategies be developed. Mouse models provide an important platform for anti-filarial drug research in a preclinical setting. This review presents an overview about the Litomosoides sigmodontis and Acanthocheilonema viteae filarial mouse models and their role in immunological research as well as preclinical studies about novel anti-filarial drugs and treatment strategies.
Collapse
Affiliation(s)
- Frederic Risch
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, Bonn, Germany
| | - Manuel Ritter
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, Bonn, Germany
| | - Achim Hoerauf
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), partner site Bonn-Cologne, Bonn, Germany
| | - Marc P Hübner
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, Bonn, Germany.
- German Center for Infection Research (DZIF), partner site Bonn-Cologne, Bonn, Germany.
| |
Collapse
|
22
|
Sibi JM, Mohan V, Munisankar S, Babu S, Aravindhan V. Augmented Innate and Adaptive Immune Responses Under Conditions of Diabetes-Filariasis Comorbidity. Front Immunol 2021; 12:716515. [PMID: 34566972 PMCID: PMC8462934 DOI: 10.3389/fimmu.2021.716515] [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: 05/28/2021] [Accepted: 08/23/2021] [Indexed: 11/13/2022] Open
Abstract
Metainflammation, as seen in chronic diabetes subjects, impairs immunity and increases the susceptibility to infections. In the present study, the effect of diabetes on immune response against filariasis was studied. Both toll-like receptor (TLR)-mediated and crude antigen-induced immune responses were quantified, in whole blood cultures from filariasis-infected subjects (LF+), with and without diabetes. Blood cultures were stimulated with TLR ligands (TLR2 and TLR4) or filarial antigen or were left unstimulated (control) for 18 h. Cytokine, chemokine, and defensin secretion was quantified by ELISA. Expression of HLA-DR, B7-1, B7-2, activation marker (CD69), and Th (Th1, Th2, Th17, and Th9) phenotypes was quantified by flow cytometry. Expression of immunomodulatory effectors (Cox-2, HO-1, IDO-1, and p47Phox) and Th-polarizing transcription factors (T-bet, GATA3, and ROR-γt) was quantified by quantitative PCR. Secretion of IL-27, IL-1Ra, IL-12, IL-33, IL-9, and SDF-1 was increased under diabetes conditions with increased Th9 polarization and increased expression of Cox-2 and IDO. Overall, diabetes was found to augment both TLR-mediated and antigen-induced inflammation, which can promote chronic pathology in LF+ subjects.
Collapse
Affiliation(s)
- Joy Manohar Sibi
- Department of Genetics, Dr A. L. Mudaliar Post Graduate Institute of Basic Medical Sciences (ALM PG IBMS), University of Madras, Chennai, India
| | - Viswanathan Mohan
- Madras Diabetes Research Foundation and Dr. Mohan's Diabetes Specialties Centre, Chennai, India
| | - Saravanan Munisankar
- National Institute of Health-International Centre for Excellence in Research, National Institute for Research in Tuberculosis, Chennai, India
| | - Subash Babu
- National Institute of Health-International Centre for Excellence in Research, National Institute for Research in Tuberculosis, Chennai, India
| | - Vivekanandhan Aravindhan
- Department of Genetics, Dr A. L. Mudaliar Post Graduate Institute of Basic Medical Sciences (ALM PG IBMS), University of Madras, Chennai, India
| |
Collapse
|
23
|
Helminth protection against type-1 diabetes: an insight into immunomodulatory effect of helminth-induced infection. Mol Biol Rep 2021; 48:6581-6588. [PMID: 34432219 DOI: 10.1007/s11033-021-06663-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 08/17/2021] [Indexed: 10/20/2022]
Abstract
Helminths are the old dirty friends of humans from decades and may live undetected by the immune system for years in the tissues. They have evolved as good experts at subverting the immune system. Despite of their pathogenicity, they provide protection to their host against certain inflammatory diseases such as diabetes by modulating the immune mechanisms. These parasites are extra-cellular and induce Th2 response which triggers the adaptive immune cells as well as innate immune cells to work synergistically allowing Tregs to work in a toll-like receptor-dependent manure. T-helper cells type-2 also secrete certain anti-inflammatory cytokines including IL-4, IL-10, IL-13 and TGF-β which also provide protection against type-1 diabetes. Several helminths such as T. crassiceps, S. venezuelensis, filarial worms, Schistosoma spp. and T. spiralis have been reported to prevent diabetes in mouse models as well as in some clinical trials. Immunomodulatory talent of helminths is receiving greater attention to prevent diabetes. Herein, an attempt has been made to review and highlight the possible immuno-modulatory mechanisms by which helminths provide protection against diabetes. Moreover, this review also emphasizes on the use of helminth-derived molecules or synthetic derivatives of helminth-antigens in clinical trials to overcome rapidly growing autoimmune disorders including diabetes.
Collapse
|
24
|
Gao YR, Zhang RH, Li R, Tang CL, Pan Q, Pen P. The effects of helminth infections against type 2 diabetes. Parasitol Res 2021; 120:1935-1942. [PMID: 34002262 DOI: 10.1007/s00436-021-07189-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 05/07/2021] [Indexed: 12/15/2022]
Abstract
Type 2 diabetes mellitus (T2D) is a prevalent inflammation-related disease characterized by insulin resistance and elevated blood glucose levels. The high incidence rate of T2D in Western societies may be due to environmental conditions, including reduced worm exposure. In human and animal models, some helminths, such as Schistosoma, Nippostrongylus, Strongyloides, and Heligmosomoides, and their products reportedly ameliorate or prevent T2D progression. T2D induces adaptive immune pathways involved in the inhibition of type 1 immune responses, promotion of type 2 immune responses, and expansion of regulatory T cells and innate immune cells, such as macrophages, eosinophils, and group 2 innate lymphoid cells. Among immune cells expanded in T2DM, type 2 immune cells and macrophages are the most important and may have synergistic effects. The stimulation of host immunity by helminth infections also promotes interactions between the innate and adaptive immune systems. In this paper, we provide a comprehensive review of intestinal helminths' protective effects against T2D.
Collapse
Affiliation(s)
- Yan-Ru Gao
- Medical Department, City College, Wuhan University of Science and Technology, Wuhan, 430083, China
| | - Rong-Hui Zhang
- Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, 430063, China
| | - Ru Li
- Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, 430063, China
| | - Chun-Lian Tang
- Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, 430063, China.
| | - Qun Pan
- Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, 430063, China.
| | - Peng Pen
- Wuhan Institute for Tuberculosis Control, Wuhan Pulmonary Hospital, Wuhan, 430030, China.
| |
Collapse
|
25
|
Dwaib HS, AlZaim I, Eid AH, Obeid O, El-Yazbi AF. Modulatory Effect of Intermittent Fasting on Adipose Tissue Inflammation: Amelioration of Cardiovascular Dysfunction in Early Metabolic Impairment. Front Pharmacol 2021; 12:626313. [PMID: 33897419 PMCID: PMC8062864 DOI: 10.3389/fphar.2021.626313] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 02/18/2021] [Indexed: 12/15/2022] Open
Abstract
Cardiometabolic syndrome (CMS) is a cluster of maladaptive cardiovascular, renal, thrombotic, inflammatory, and metabolic disorders. It confers a high risk of cardiovascular mortality and morbidity. CMS is triggered by major shifts in lifestyle and dietary habits with increased consumption of refined, calorie-dense diets. Evidence indicates that diet-induced CMS is linked to Adipose tissue (AT) inflammation. This led to the proposal that adipose inflammation may be involved in metabolic derangements, such as insulin resistance and poor glycemic control, as well as the contribution to the inflammatory process predisposing patients to increased cardiovascular risk. Therefore, in the absence of direct pharmacological interventions for the subclinical phase of CMS, time restricted feeding regimens were anticipated to alleviate early metabolic damage and subsequent comorbidities. These regimens, referred to as intermittent fasting (IF), showed a strong positive impact on the metabolic state of obese and non-obese human subjects and animal models, positive AT remodeling in face of overnutrition and high fat diet (HFD) consumption, and improved CV outcomes. Here, we summarize the available evidence on the role of adipose inflammation in triggering cardiovascular impairment in the context of diet induced CMS with an emphasis on the involvement of perivascular adipose tissue. As well, we propose some possible molecular pathways linking intermittent fasting to the ameliorative effect on adipose inflammation and cardiovascular dysfunction under such circumstances. We highlight a number of targets, whose function changes in perivascular adipose tissue inflammation and could be modified by intermittent fasting acting as a novel approach to ameliorate the inflammatory status.
Collapse
Affiliation(s)
- Haneen S. Dwaib
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences, American University of Beirut, Beirut, Lebanon
| | - Ibrahim AlZaim
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ali H. Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Omar Obeid
- Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences, American University of Beirut, Beirut, Lebanon
| | - Ahmed F. El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- Faculty of Pharmacy, Al-Alamein International University, Alamein, Egypt
| |
Collapse
|
26
|
Rennie C, Fernandez R, Donnelly S, McGrath KCY. The Impact of Helminth Infection on the Incidence of Metabolic Syndrome: A Systematic Review and Meta-Analysis. Front Endocrinol (Lausanne) 2021; 12:728396. [PMID: 34456879 PMCID: PMC8397462 DOI: 10.3389/fendo.2021.728396] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 07/20/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND There are a growing number of publications that report an absence of inflammatory based disease among populations that are endemic to parasitic worms (helminths) demonstrating the ability of these parasites to potentially regulate human immune responses. The aim of this systematic review and meta-analysis was to determine the impact of helminth infection on metabolic outcomes in human populations. METHODS Using PRISMA guidelines, six databases were searched for studies published up to August 2020. Random effects meta-analysis was performed to estimate pooled proportions with 95% confidence intervals using the Review Manager Software version 5.4.1. RESULTS Fourteen studies were included in the review. Fasting blood glucose was significantly lower in persons with infection (MD -0.22, 95% CI -0.40- -0.04, P=0.02), HbA1c levels were lower, although not significantly, and prevalence of the metabolic syndrome (P=0.001) and type 2 diabetes was lower (OR 1.03, 95% CI 0.34-3.09, P<0.0001). Infection was negatively associated with type 2 diabetes when comparing person with diabetes to the group without diabetes (OR 0.44, 95% CI 0.29-0.67, P=0.0001). CONCLUSIONS While infection with helminths was generally associated with improved metabolic function, there were notable differences in efficacy between parasite species. Based on the data assessed, live infection with S. mansoni resulted in the most significant positive changes to metabolic outcomes. SYSTEMATIC REVIEW REGISTRATION Website: PROSPERO Identified: CRD42021227619.
Collapse
Affiliation(s)
- Claire Rennie
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Ritin Fernandez
- School of Nursing, University of Wollongong, Wollongong, NSW, Australia
- Centre for Research in Nursing and Health, St George Hospital, Sydney, NSW, Australia
- Centre for Evidence Based Initiatives in Health Care a JBI Centre of Excellence, Sydney, NSW, Australia
| | - Sheila Donnelly
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
- *Correspondence: Kristine CY McGrath, ; Sheila Donnelly,
| | - Kristine CY McGrath
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
- *Correspondence: Kristine CY McGrath, ; Sheila Donnelly,
| |
Collapse
|
27
|
Vanhamme L, Souopgui J, Ghogomu S, Ngale Njume F. The Functional Parasitic Worm Secretome: Mapping the Place of Onchocerca volvulus Excretory Secretory Products. Pathogens 2020; 9:pathogens9110975. [PMID: 33238479 PMCID: PMC7709020 DOI: 10.3390/pathogens9110975] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 01/15/2023] Open
Abstract
Nematodes constitute a very successful phylum, especially in terms of parasitism. Inside their mammalian hosts, parasitic nematodes mainly dwell in the digestive tract (geohelminths) or in the vascular system (filariae). One of their main characteristics is their long sojourn inside the body where they are accessible to the immune system. Several strategies are used by parasites in order to counteract the immune attacks. One of them is the expression of molecules interfering with the function of the immune system. Excretory-secretory products (ESPs) pertain to this category. This is, however, not their only biological function, as they seem also involved in other mechanisms such as pathogenicity or parasitic cycle (molting, for example). We will mainly focus on filariae ESPs with an emphasis on data available regarding Onchocerca volvulus, but we will also refer to a few relevant/illustrative examples related to other worm categories when necessary (geohelminth nematodes, trematodes or cestodes). We first present Onchocerca volvulus, mainly focusing on the aspects of this organism that seem relevant when it comes to ESPs: life cycle, manifestations of the sickness, immunosuppression, diagnosis and treatment. We then elaborate on the function and use of ESPs in these aspects.
Collapse
Affiliation(s)
- Luc Vanhamme
- Department of Molecular Biology, Institute of Biology and Molecular Medicine, IBMM, Université Libre de Bruxelles, Rue des Professeurs Jeener et Brachet 12, 6041 Gosselies, Belgium; (J.S.); (F.N.N.)
- Correspondence:
| | - Jacob Souopgui
- Department of Molecular Biology, Institute of Biology and Molecular Medicine, IBMM, Université Libre de Bruxelles, Rue des Professeurs Jeener et Brachet 12, 6041 Gosselies, Belgium; (J.S.); (F.N.N.)
| | - Stephen Ghogomu
- Molecular and Cell Biology Laboratory, Biotechnology Unit, University of Buea, Buea P.O Box 63, Cameroon;
| | - Ferdinand Ngale Njume
- Department of Molecular Biology, Institute of Biology and Molecular Medicine, IBMM, Université Libre de Bruxelles, Rue des Professeurs Jeener et Brachet 12, 6041 Gosselies, Belgium; (J.S.); (F.N.N.)
- Molecular and Cell Biology Laboratory, Biotechnology Unit, University of Buea, Buea P.O Box 63, Cameroon;
| |
Collapse
|
28
|
AlZaim I, Hammoud SH, Al-Koussa H, Ghazi A, Eid AH, El-Yazbi AF. Adipose Tissue Immunomodulation: A Novel Therapeutic Approach in Cardiovascular and Metabolic Diseases. Front Cardiovasc Med 2020; 7:602088. [PMID: 33282920 PMCID: PMC7705180 DOI: 10.3389/fcvm.2020.602088] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/22/2020] [Indexed: 12/12/2022] Open
Abstract
Adipose tissue is a critical regulator of systemic metabolism and bodily homeostasis as it secretes a myriad of adipokines, including inflammatory and anti-inflammatory cytokines. As the main storage pool of lipids, subcutaneous and visceral adipose tissues undergo marked hypertrophy and hyperplasia in response to nutritional excess leading to hypoxia, adipokine dysregulation, and subsequent low-grade inflammation that is characterized by increased infiltration and activation of innate and adaptive immune cells. The specific localization, physiology, susceptibility to inflammation and the heterogeneity of the inflammatory cell population of each adipose depot are unique and thus dictate the possible complications of adipose tissue chronic inflammation. Several lines of evidence link visceral and particularly perivascular, pericardial, and perirenal adipose tissue inflammation to the development of metabolic syndrome, insulin resistance, type 2 diabetes and cardiovascular diseases. In addition to the implication of the immune system in the regulation of adipose tissue function, adipose tissue immune components are pivotal in detrimental or otherwise favorable adipose tissue remodeling and thermogenesis. Adipose tissue resident and infiltrating immune cells undergo metabolic and morphological adaptation based on the systemic energy status and thus a better comprehension of the metabolic regulation of immune cells in adipose tissues is pivotal to address complications of chronic adipose tissue inflammation. In this review, we discuss the role of adipose innate and adaptive immune cells across various physiological and pathophysiological states that pertain to the development or progression of cardiovascular diseases associated with metabolic disorders. Understanding such mechanisms allows for the exploitation of the adipose tissue-immune system crosstalk, exploring how the adipose immune system might be targeted as a strategy to treat cardiovascular derangements associated with metabolic dysfunctions.
Collapse
Affiliation(s)
- Ibrahim AlZaim
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Safaa H. Hammoud
- Department of Pharmacology and Therapeutics, Beirut Arab University, Beirut, Lebanon
| | - Houssam Al-Koussa
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon
| | - Alaa Ghazi
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon
| | - Ali H. Eid
- Department of Pharmacology and Therapeutics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Basic Medical Sciences, College of Medicine, Qatar University, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Ahmed F. El-Yazbi
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
29
|
Rajamanickam A, Munisankar S, Thiruvengadam K, Menon PA, Dolla C, Nutman TB, Babu S. Impact of Helminth Infection on Metabolic and Immune Homeostasis in Non-diabetic Obesity. Front Immunol 2020; 11:2195. [PMID: 33042134 PMCID: PMC7524873 DOI: 10.3389/fimmu.2020.02195] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 08/11/2020] [Indexed: 12/11/2022] Open
Abstract
Several epidemiological and immunological studies indicate a reciprocal association between obesity/metabolic syndrome and helminth infections. Numerous studies demonstrated that obesity is concomitant with chronic low-grade inflammation, which is marked by vital changes in cellular composition and function of adipose tissue. However, the effect of helminth infection on the homeostatic milieu in obesity is not well-understood. To determine the relationship between Strongyloides stercoralis (Ss) infection and obesity, we examined an array of parameters linked with obesity both before and at 6 months following anthelmintic treatment. To this end, we measured serum levels of pancreatic hormones, incretins, adipokines and Type-1, Type-2, Type-17, and other proinflammatory cytokines in those with non-diabetic obesity with (INF) or without Ss infection (UN). In INF individuals, we evaluated the levels of these parameters at 6 months following anthelmintic treatment. INF individuals revealed significantly lower levels of insulin, glucagon, C-peptide, and GLP-1 and significantly elevated levels of GIP compared to UN individuals. INF individuals also showed significantly lower levels of Type-1, Type-17 and other pro-inflammatory cytokines and significantly increased levels of Type-2 and regulatory cytokines in comparison to UN individuals. Most of these changes were significantly reversed following anthelmintic treatment. Ss infection is associated with a significant alteration of pancreatic hormones, incretins, adipokines, and cytokines in obese individuals and its partial reversal following anthelmintic treatment. Our data offer a possible biological mechanism for the protective effect of Ss infection on obesity.
Collapse
Affiliation(s)
- Anuradha Rajamanickam
- National Institute of Health-National Institute for Research in Tuberculosis (NIRT)-International Center for Excellence in Research, Chennai, India
| | - Saravanan Munisankar
- National Institute of Health-National Institute for Research in Tuberculosis (NIRT)-International Center for Excellence in Research, Chennai, India
| | - Kannan Thiruvengadam
- Department of Epidemiology, National Institute for Research in Tuberculosis, Chennai, India
| | - Pradeep A Menon
- Department of Epidemiology, National Institute for Research in Tuberculosis, Chennai, India
| | - Chandrakumar Dolla
- Department of Epidemiology, National Institute for Research in Tuberculosis, Chennai, India
| | - Thomas B Nutman
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Subash Babu
- National Institute of Health-National Institute for Research in Tuberculosis (NIRT)-International Center for Excellence in Research, Chennai, India.,Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States.,Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, Frederick, MD, United States
| |
Collapse
|
30
|
Vohralik EJ, Psaila AM, Knights AJ, Quinlan KGR. EoTHINophils: Eosinophils as key players in adipose tissue homeostasis. Clin Exp Pharmacol Physiol 2020; 47:1495-1505. [PMID: 32163614 DOI: 10.1111/1440-1681.13304] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 02/24/2020] [Accepted: 03/09/2020] [Indexed: 12/22/2022]
Abstract
Eosinophils are granular cells of the innate immune system that are found in almost all vertebrates and some invertebrates. Knowledge of their wide-ranging roles in health and disease has largely been attained through studies in mice and humans. Although eosinophils are typically associated with helminth infections and allergic diseases such as asthma, there is building evidence that beneficial homeostatic eosinophils residing in specific niches are important for tissue development, remodelling and metabolic control. In recent years, the importance of immune cells in the regulation of adipose tissue homeostasis has been a focal point of research efforts. There is an abundance of anti-inflammatory innate immune cells in lean white adipose tissue, including macrophages, eosinophils and group 2 innate lymphoid cells, which promote energy homeostasis and stimulate the development of thermogenic beige adipocytes. This review will evaluate evidence for the role of adipose-resident eosinophils in local tissue homeostasis, beiging and systemic metabolism, highlighting where more research is needed to establish the specific effector functions that adipose eosinophils perform in response to different internal and external cues.
Collapse
Affiliation(s)
- Emily J Vohralik
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Annalise M Psaila
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Alexander J Knights
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Kate G R Quinlan
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|
31
|
Sanya RE, Webb EL, Zziwa C, Kizindo R, Sewankambo M, Tumusiime J, Nakazibwe E, Oduru G, Niwagaba E, Nakawungu PK, Kabagenyi J, Nassuuna J, Walusimbi B, Andia-Biraro I, Elliott AM. The Effect of Helminth Infections and Their Treatment on Metabolic Outcomes: Results of a Cluster-Randomized Trial. Clin Infect Dis 2020; 71:601-613. [PMID: 31504336 PMCID: PMC7384320 DOI: 10.1093/cid/ciz859] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 08/28/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Helminths may protect against cardiometabolic risk through effects on inflammation and metabolism; their treatment may be detrimental to metabolic outcomes. METHODS In a cluster-randomized trial in 26 Ugandan fishing communities we investigated effects of community-wide intensive (quarterly single-dose praziquantel, triple-dose albendazole) vs standard (annual single-dose praziquantel, biannual single-dose albendazole) anthelminthic treatment on metabolic outcomes, and observational associations between helminths and metabolic outcomes. The primary outcome, homeostatic model assessment of insulin resistance (HOMA-IR), and secondary outcomes (including blood pressure, fasting blood glucose, lipids) were assessed after 4 years' intervention among individuals aged ≥10 years. RESULTS We analyzed 1898 participants. Intensive treatment had no effect on HOMA-IR (adjusted geometric mean ratio, 0.96 [95% confidence interval {CI}, .86-1.07]; P = .42) but resulted in higher mean low-density lipoprotein cholesterol (LDL-c) (2.86 vs 2.60 mmol/L; adjusted mean difference, 0.26 [95% CI, -.03 to .56]; P = .08). Lower LDL-c levels were associated with Schistosoma mansoni (2.37 vs 2.80 mmol/L; -0.25 [95% CI, -.49 to -.02]; P = .04) or Strongyloides (2.34 vs 2.69 mmol/L; -0.32 [95% CI, -.53 to -.12]; P = .003) infection. Schistosoma mansoni was associated with lower total cholesterol (4.24 vs 4.64 mmol/L; -0.25 [95% CI, -.44 to -.07]; P = .01) and moderate to heavy S. mansoni infection with lower triglycerides, LDL-c, and diastolic blood pressure. CONCLUSIONS Helminth infections improve lipid profiles and may lower blood pressure. Studies to confirm causality and investigate mechanisms may contribute to understanding the epidemiological transition and suggest new approaches to prevent cardiometabolic disease. CLINICAL TRIALS REGISTRATION ISRCTN47196031.
Collapse
Affiliation(s)
- Richard E Sanya
- Immunomodulation and Vaccines Programme, Medical Research Council (MRC)/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- Department of Internal Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Emily L Webb
- MRC Tropical Epidemiology Group, Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Christopher Zziwa
- Immunomodulation and Vaccines Programme, Medical Research Council (MRC)/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Robert Kizindo
- Immunomodulation and Vaccines Programme, Medical Research Council (MRC)/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Moses Sewankambo
- Immunomodulation and Vaccines Programme, Medical Research Council (MRC)/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Josephine Tumusiime
- Immunomodulation and Vaccines Programme, Medical Research Council (MRC)/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Esther Nakazibwe
- Immunomodulation and Vaccines Programme, Medical Research Council (MRC)/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Gloria Oduru
- Immunomodulation and Vaccines Programme, Medical Research Council (MRC)/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Emmanuel Niwagaba
- Immunomodulation and Vaccines Programme, Medical Research Council (MRC)/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Prossy Kabuubi Nakawungu
- Immunomodulation and Vaccines Programme, Medical Research Council (MRC)/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Joyce Kabagenyi
- Immunomodulation and Vaccines Programme, Medical Research Council (MRC)/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Jacent Nassuuna
- Immunomodulation and Vaccines Programme, Medical Research Council (MRC)/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Bridgious Walusimbi
- Immunomodulation and Vaccines Programme, Medical Research Council (MRC)/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Irene Andia-Biraro
- Immunomodulation and Vaccines Programme, Medical Research Council (MRC)/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- Department of Internal Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Alison M Elliott
- Immunomodulation and Vaccines Programme, Medical Research Council (MRC)/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
32
|
Finlay CM, Allen JE. The immune response of inbred laboratory mice to Litomosoides sigmodontis: A route to discovery in myeloid cell biology. Parasite Immunol 2020; 42:e12708. [PMID: 32145033 PMCID: PMC7317388 DOI: 10.1111/pim.12708] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/12/2020] [Accepted: 02/24/2020] [Indexed: 12/11/2022]
Abstract
Litomosoides sigmodontis is the only filarial nematode where the full life cycle, from larval delivery to the skin through to circulating microfilaria, can be completed in immunocompetent laboratory mice. It is thus an invaluable tool for the study of filariasis. It has been used for the study of novel anti-helminthic therapeutics, the development of vaccines against filariasis, the development of immunomodulatory drugs for the treatment of inflammatory disease and the study of basic immune responses to filarial nematodes. This review will focus on the latter and aims to summarize how the L sigmodontis model has advanced our basic understanding of immune responses to helminths, led to major discoveries in macrophage biology and provided new insights into the immunological functions of the pleural cavity. Finally, and most importantly L sigmodontis represents a suitable platform to study how host genotype affects immune responses, with the potential for further discovery in myeloid cell biology and beyond.
Collapse
Affiliation(s)
- Conor M Finlay
- Lydia Becker Institute for Immunology & Infection, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Judith E Allen
- Lydia Becker Institute for Immunology & Infection, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| |
Collapse
|
33
|
Crowe J, Lumb FE, Doonan J, Broussard M, Tarafdar A, Pineda MA, Landabaso C, Mulvey L, Hoskisson PA, Babayan SA, Selman C, Harnett W, Harnett MM. The parasitic worm product ES-62 promotes health- and life-span in a high calorie diet-accelerated mouse model of ageing. PLoS Pathog 2020; 16:e1008391. [PMID: 32163524 PMCID: PMC7108737 DOI: 10.1371/journal.ppat.1008391] [Citation(s) in RCA: 24] [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: 08/09/2019] [Revised: 03/31/2020] [Accepted: 02/07/2020] [Indexed: 12/15/2022] Open
Abstract
Improvements in hygiene and health management have driven significant increases in human lifespan over the last 50 years. Frustratingly however, this extension of lifespan has not been matched by equivalent improvements in late-life health, not least due to the global pandemic in type-2 diabetes, obesity and cardiovascular disease, all ageing-associated conditions exacerbated and accelerated by widespread adoption of the high calorie Western diet (HCD). Recently, evidence has begun to emerge that parasitic worm infection might protect against such ageing-associated co-morbidities, as a serendipitous side-effect of their evolution of pro-survival, anti-inflammatory mechanisms. As a novel therapeutic strategy, we have therefore investigated the potential of ES-62, an anti-inflammatory secreted product of the filarial nematode Acanthocheilonema viteae, to improve healthspan (the period of life before diseases of ageing appear) by targeting the chronic inflammation that drives metabolic dysregulation underpinning ageing-induced ill-health. We administered ES-62 subcutaneously (at a dose of 1 μg/week) to C57BL/6J mice undergoing HCD-accelerated ageing throughout their lifespan, while subjecting the animals to analysis of 120 immunometabolic responses at various time-points. ES-62 improved a number of inflammatory parameters, but markedly, a range of pathophysiological, metabolic and microbiome parameters of ageing were also successfully targeted. Notably, ES-62-mediated promotion of healthspan in male and female HCD-mice was associated with different mechanisms and reflecting this, machine learning modelling identified sex-specific signatures predictive of ES-62 action against HCD-accelerated ageing. Remarkably, ES-62 substantially increased the median survival of male HCD-mice. This was not the case with female animals and unexpectedly, this difference between the two sexes could not be explained in terms of suppression of the chronic inflammation driving ageing, as ES-62 tended to be more effective in reducing this in female mice. Rather, the difference appeared to be associated with ES-62's additional ability to preferentially promote a healthier gut-metabolic tissue axis in male animals.
Collapse
Affiliation(s)
- Jenny Crowe
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Felicity E. Lumb
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - James Doonan
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Margaux Broussard
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Anuradha Tarafdar
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Miguel A. Pineda
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Carmen Landabaso
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Lorna Mulvey
- Glasgow Ageing Research Network (GARNER), Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, United Kingdom
| | - Paul A. Hoskisson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Simon A. Babayan
- Glasgow Ageing Research Network (GARNER), Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, United Kingdom
| | - Colin Selman
- Glasgow Ageing Research Network (GARNER), Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, United Kingdom
| | - William Harnett
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Margaret M. Harnett
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
34
|
Frohberger SJ, Fercoq F, Neumann AL, Surendar J, Stamminger W, Ehrens A, Karunakaran I, Remion E, Vogl T, Hoerauf A, Martin C, Hübner MP. S100A8/S100A9 deficiency increases neutrophil activation and protective immune responses against invading infective L3 larvae of the filarial nematode Litomosoides sigmodontis. PLoS Negl Trop Dis 2020; 14:e0008119. [PMID: 32107497 PMCID: PMC7064255 DOI: 10.1371/journal.pntd.0008119] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 03/10/2020] [Accepted: 02/05/2020] [Indexed: 12/17/2022] Open
Abstract
Neutrophils are essentially involved in protective immune responses against invading infective larvae of filarial nematodes. The present study investigated the impact of S100A8/S100A9 on protective immune responses against the rodent filarial nematode Litomosoides sigmodontis. S100A9 forms with S100A8 the heterodimer calprotectin, which is expressed by circulating neutrophils and monocytes and mitigates or amplifies tissue damage as well as inflammation depending on the immune environment. Mice deficient for S100A8/A9 had a significantly reduced worm burden in comparison to wildtype (WT) animals 12 days after infection (dpi) with infective L3 larvae, either by the vector or subcutaneous inoculation, the latter suggesting that circumventing natural immune responses within the epidermis and dermis do not alter the phenotype. Nevertheless, upon intradermal injection of L3 larvae, increased total numbers of neutrophils, eosinophils and macrophages were observed within the skin of S100A8/A9-/- mice. Furthermore, upon infection the bronchoalveolar and thoracic cavity lavage of S100A8/A9-/- mice showed increased concentrations of CXCL-1, CXCL-2, CXCL-5, as well as elastase in comparison to the WT controls. Neutrophils from S100A8/A9-/- mice exhibited an increased in vitro activation and reduced L3 larval motility more effectively in vitro compared to WT neutrophils. The depletion of neutrophils from S100A8/A9-/- mice prior to L. sigmodontis infection until 5dpi abrogated the protective effect and led to an increased worm burden, indicating that neutrophils mediate enhanced protective immune responses against invading L3 larvae in S100A8/A9-/- mice. Interestingly, complete circumvention of protective immune responses in the skin and the lymphatics by intravenous injection of L3 larvae reversed the phenotype and resulted in an increased worm burden in S100A8/A9-/- mice. In summary, our results reveal that lack of S100A8/S100A9 triggers L3-induced inflammatory responses, increasing chemokine levels, granulocyte recruitment as well as neutrophil activation and therefore impairs larval migration and susceptibility for filarial infection.
Collapse
Affiliation(s)
- Stefan J. Frohberger
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Frederic Fercoq
- Unité Molécules de Communication et Adaptation des Microorganismes (MCAM, UMR 7245), Muséum national d’Histoire naturelle, CNRS; Paris, France
| | - Anna-Lena Neumann
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Jayagopi Surendar
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Wiebke Stamminger
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Alexandra Ehrens
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Indulekha Karunakaran
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Estelle Remion
- Unité Molécules de Communication et Adaptation des Microorganismes (MCAM, UMR 7245), Muséum national d’Histoire naturelle, CNRS; Paris, France
| | - Thomas Vogl
- Institute of Immunology, University Hospital of Münster, Münster, Germany
| | - Achim Hoerauf
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), partner site Bonn-Cologne, Bonn, Germany
| | - Coralie Martin
- Unité Molécules de Communication et Adaptation des Microorganismes (MCAM, UMR 7245), Muséum national d’Histoire naturelle, CNRS; Paris, France
| | - Marc P. Hübner
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
- * E-mail:
| |
Collapse
|
35
|
Berbudi A, Rahmadika N, Tjahjadi AI, Ruslami R. Type 2 Diabetes and its Impact on the Immune System. Curr Diabetes Rev 2020; 16:442-449. [PMID: 31657690 PMCID: PMC7475801 DOI: 10.2174/1573399815666191024085838] [Citation(s) in RCA: 500] [Impact Index Per Article: 100.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 07/24/2019] [Accepted: 09/16/2019] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Type 2 Diabetes (T2D) is a major health problem worldwide. This metabolic disease is indicated by high blood glucose levels due to insufficient insulin production by the pancreas. An inflammatory response occurs as a result of the immune response to high blood glucose levels as well as the presence of inflammatory mediators produced by adipocytes and macrophages in fat tissue. This low and chronic inflammation damages the pancreatic beta cells and leads to insufficient insulin production, which results in hyperglycemia. Hyperglycemia in diabetes is thought to cause dysfunction of the immune response, which fails to control the spread of invading pathogens in diabetic subjects. Therefore, diabetic subjects are known to more susceptible to infections. The increased prevalence of T2D will increase the incidence of infectious diseases and related comorbidities. OBJECTIVE This review provides an overview of the immunological aspect of T2D and the possible mechanisms that result in increased infections in diabetics. CONCLUSION A better understanding of how immune dysfunctions occur during hyperglycemia can lead to novel treatments and preventions for infectious diseases and T2D comorbidities, thus improving the outcome of infectious disease treatment in T2D patients.
Collapse
Affiliation(s)
- Afiat Berbudi
- Department of Biomedical Sciences, Parasitology Division, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
- Infectious Disease Research Center, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
- Address correspondence to this author at the Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Jl. Raya Bandung Sumedang Km. 21, 45363, West Java, Indonesia; E-mail:
| | - Nofri Rahmadika
- Infectious Disease Research Center, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Adi Imam Tjahjadi
- Infectious Disease Research Center, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
- Department of Biomedical Sciences, Microbiology Division, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Rovina Ruslami
- Infectious Disease Research Center, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
- Department of Biomedical Sciences, Pharmacology and Therapy Division, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| |
Collapse
|
36
|
Surendar J, Karunakaran I, Frohberger SJ, Koschel M, Hoerauf A, Hübner MP. Macrophages Mediate Increased CD8 T Cell Inflammation During Weight Loss in Formerly Obese Mice. Front Endocrinol (Lausanne) 2020; 11:257. [PMID: 32411095 PMCID: PMC7198814 DOI: 10.3389/fendo.2020.00257] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 04/06/2020] [Indexed: 12/27/2022] Open
Abstract
Even after successful weight reduction, obese adults tend to quickly regain the lost weight. This raises the question of whether weight loss improves the underlying chronic adipose tissue inflammation characteristic of obesity. In order to improve our understanding of the mechanisms that reshape metabolic organs during weight loss, we investigated the macrophage and T cell function of the liver and adipose tissue on reversing high fat diet (HFD) mice to normal control diet (NCD). Obese mice that were switched to NCD showed an improvement in their metabolic profile that included enhanced glucose and insulin tolerance, decreased cholesterol, triglyceride, serum glutamic-oxaloacetic transaminase (SGOT), and serum glutamic pyruvic transaminase (SGPT) levels that were comparable to NCD controls. However, despite weight loss, increased frequencies, but not total numbers, of IL-17+ and IL-22+ CD4+ T cells, IFN-γ+ and TNF+ CD8+ T cells and IL-17+ and IL-22+ CD8+ T cells were observed in the adipose tissue of mice switched from HFD to NCD compared to NCD and even HFD fed mice. Further, in the liver, IFN-γ+ and TNF+ CD8+ T cell, IL-17+ and IL-22+ CD8+ T cell, macrophage frequencies and their expression of antigen presenting molecules were increased. To determine if macrophages are the major determinants of the sustained inflammation observed during weight loss, we depleted macrophages, which significantly reduced IFN-γ+, TNF+, IL-17+, and IL-22+ CD8+ T cell frequencies in the liver and the adipose tissue. In conclusion, we show that although weight loss improves the metabolic profile, there is an active and ongoing CD8+ T cell inflammation in liver and adipose tissue mediated by macrophages.
Collapse
Affiliation(s)
- Jayagopi Surendar
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Indulekha Karunakaran
- 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
| | - Marianne Koschel
- 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 Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| | - Marc P. Hübner
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
- *Correspondence: Marc P. Hübner
| |
Collapse
|
37
|
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: 17] [Impact Index Per Article: 3.4] [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
|
38
|
Surendar J, Frohberger SJ, Karunakaran I, Schmitt V, Stamminger W, Neumann AL, Wilhelm C, Hoerauf A, Hübner MP. Adiponectin Limits IFN-γ and IL-17 Producing CD4 T Cells in Obesity by Restraining Cell Intrinsic Glycolysis. Front Immunol 2019; 10:2555. [PMID: 31736971 PMCID: PMC6828851 DOI: 10.3389/fimmu.2019.02555] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 10/15/2019] [Indexed: 12/13/2022] Open
Abstract
Compared to the innate immune system, the contribution of the adaptive immune response during obesity and insulin resistance is still not completely understood. Here we demonstrate that high fat diet (HFD) increases the frequencies of activated CD4+ and CD8+ T cells and frequencies of T cells positive for IFN-γ and IL-17 in the adipose tissue. The adipocyte-derived soluble factor adiponectin reduces IFN-γ and IL-17 positive CD4+ T cells from HFD mice and dampens the differentiation of naïve T cells into Th1 cells and Th17 cells. Adiponectin reduces Th17 cell differentiation and restrains glycolysis in an AMPK dependent fashion. Treatment with adult worm extracts of the rodent filarial nematode Litomosoides sigmodontis (LsAg) reduces adipose tissue Th1 and Th17 cell frequencies during HFD and increases adiponectin levels. Stimulation of T cells in the presence of adipocyte-conditioned media (ACM) from LsAg-treated mice reduces Th1 and Th17 frequencies and this effect was abolished when ACM was treated with an adiponectin neutralizing antibody. Collectively, these data reveal a novel role of adiponectin in controlling pro-inflammatory CD4+ T cells during obesity and suggest that the beneficial role of helminth infections and helminth-derived products on obesity and insulin resistance may be in part mediated by adiponectin.
Collapse
Affiliation(s)
- Jayagopi Surendar
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany.,Unit for Immunopathology, Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Stefan J Frohberger
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Indulekha Karunakaran
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Vanessa Schmitt
- Unit for Immunopathology, Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Wiebke Stamminger
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Anna-Lena Neumann
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Christoph Wilhelm
- Unit for Immunopathology, Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Achim Hoerauf
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany.,German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| | - Marc P Hübner
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
39
|
Lumb FE, Crowe J, Doonan J, Suckling CJ, Selman C, Harnett MM, Harnett W. Synthetic small molecule analogues of the immunomodulatory Acanthocheilonema viteae product ES-62 promote metabolic homeostasis during obesity in a mouse model. Mol Biochem Parasitol 2019; 234:111232. [PMID: 31634505 DOI: 10.1016/j.molbiopara.2019.111232] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/08/2019] [Accepted: 10/15/2019] [Indexed: 02/06/2023]
Abstract
One of the most rapidly increasing human public health problems is obesity, whose sequelae like type-2 diabetes, represent continuously worsening, life-long conditions. Over the last 15 years, data have begun to emerge from human and more frequently, mouse studies, that support the idea that parasitic worm infection can protect against this condition. We have therefore investigated the potential of two synthetic small molecule analogues (SMAs) of the anti-inflammatory Acanthocheilonema viteae product ES-62, to protect against metabolic dysfunction in a C57BL/6 J mouse model of high calorie diet-induced obesity. We found weekly subcutaneous administration of the SMAs in combination (1 μg of each), starting one week before continuous exposure to high calorie diet (HCD), decreased fasting glucose levels and reversed the impaired glucose clearance observed in male mice, when measured at approximately 7 and 13 weeks after exposure to HCD. Fasting glucose levels were also-reduced in male mice fed a HCD for some 38 weeks when given SMA-treatment 13 weeks after the start of HCD, indicating an SMA-therapeutic potential. For the most part, protective effects were not observed in female mice. SMA treatment also conferred protection against each of reduced ileum villus length and liver fibrosis, but more prominently in female mice. Previous studies in mice indicate that protection against metabolic dysfunction is usually associated with polarisation of the immune system towards a type-2/anti-inflammatory direction but our attempts to correlate improved metabolic parameters with such changes were unsuccessful. Further analysis will therefore be required to define mechanism of action. Nevertheless, overall our data clearly show the potential of the drug-like SMAs as a preventative or treatment for metabolic dysregulation associated with obesity.
Collapse
Affiliation(s)
- Felicity E Lumb
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
| | - Jenny Crowe
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK
| | - James Doonan
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
| | - Colin J Suckling
- Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow G1 1XL, UK
| | - Colin Selman
- Glasgow Ageing Research Network (GARNER), Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow G12 8QQ, UK
| | - Margaret M Harnett
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK
| | - William Harnett
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK.
| |
Collapse
|
40
|
Bolus WR, Hasty AH. Contributions of innate type 2 inflammation to adipose function. J Lipid Res 2019; 60:1698-1709. [PMID: 29891508 PMCID: PMC6795076 DOI: 10.1194/jlr.r085993] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 05/25/2018] [Indexed: 12/17/2022] Open
Abstract
A critical contributor to the health consequences of the obesity epidemic is dysregulated adipose tissue (AT) homeostasis. While white, brown, and beige AT function is altered in obesity-related disease, white AT is marked by progressive inflammation and adipocyte dysfunction and has been the focus of extensive "immunometabolism" research in the past decade. The exact triggering events initiating and sustaining AT inflammation are still under study, but it has been shown that reducing inflammation improves insulin action in AT. Scientific efforts seeking interventions to mitigate obesity-associated AT inflammation continue, and many groups are now determining how lean healthy AT homeostasis is maintained in order to leverage these mechanisms as therapeutic targets. Such studies have revealed that an elaborate network of immune cells, cytokines, and other cellular mediators coordinate AT function. Recent studies elucidated the involvement of the innate immune system in AT homeostasis (e.g., beiging and insulin sensitivity), including M2-like macrophages, eosinophils, innate lymphoid type 2 cells, and several others. In this review, we summarize the existing literature on innate type 2 inflammation in AT; additionally, we draw attention to areas of debate where seemingly conflicting data promises to yield more surprising and elegant biology as studies continue to dissect AT physiology.
Collapse
Affiliation(s)
- W Reid Bolus
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville TN 37232
| | - Alyssa H Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville TN 37232
- Veterans Affairs Tennessee Valley Healthcare System, Nashville TN 37212
| |
Collapse
|
41
|
van der Zande HJP, Zawistowska-Deniziak A, Guigas B. Immune Regulation of Metabolic Homeostasis by Helminths and Their Molecules. Trends Parasitol 2019; 35:795-808. [PMID: 31492623 DOI: 10.1016/j.pt.2019.07.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 07/28/2019] [Accepted: 07/30/2019] [Indexed: 12/11/2022]
Abstract
Since time immemorial, humans have coevolved with a wide variety of parasitic helminths that have contributed to shape their immune system. The recent eradication of helminth infections in modern societies has coincided with a spectacular rise in inflammatory metabolic diseases, such as obesity, nonalcoholic steatohepatitis, and type 2 diabetes. Landmark studies in the emerging field of immunometabolism have highlighted the central role of the immune system in regulating metabolic functions, notably in adipose tissue, liver, and the gut. In this review we discuss how helminths, which are among the strongest natural inducers of type 2 immunity, and some of their unique immunomodulatory molecules, may contribute to the maintenance of tissue-specific and whole-body metabolic homeostasis and protection against obesity-associated meta-inflammation.
Collapse
Affiliation(s)
| | - Anna Zawistowska-Deniziak
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands; Witold Stefański Institute of Parasitology, Polish Academy of Sciences, Warsaw, Poland
| | - Bruno Guigas
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands. @lumc.nl
| |
Collapse
|
42
|
Ryan NM, Oghumu S. Role of mast cells in the generation of a T-helper type 2 dominated anti-helminthic immune response. Biosci Rep 2019; 39:BSR20181771. [PMID: 30670631 PMCID: PMC6379226 DOI: 10.1042/bsr20181771] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 01/16/2019] [Accepted: 01/21/2019] [Indexed: 12/19/2022] Open
Abstract
Mast cells are long-lived, innate immune cells of the myeloid lineage which are found in peripheral tissues located throughout the body, and positioned at the interface between the host and the environment. Mast cells are found in high concentrations during helminth infection. Using Kitw-sh mast cell deficient mice, a recently published study in Bioscience Reports by Gonzalez et al. (Biosci. Rep., 2018) focused on the role of mast cells in the immune response to infection by the helminth Hymenolepis diminuta The authors showed that mast cells play a role in the modulation of Th2 immune response characterized by a unique IL-4, IL-5 and IL-13 cytokine profile, as well as subsequent robust worm expulsion during H. diminuta infection. Unlike WT mice which expelled H. diminuta at day 10, Kitw-sh deficient mice displayed delayed worm expulsion (day 14 post infection). Further, a possible role for mast cells in the basal expression of cytokines IL-25, IL-33 and thymic stromal lymphopoietin was described. Deletion of neutrophils in Kitw-sh deficient mice enhanced H. diminuta expulsion, which was accompanied by splenomegaly. However, interactions between mast cells and other innate and adaptive immune cells during helminth infections are yet to be fully clarified. We conclude that the elucidation of mechanisms underlying mast cell interactions with cells of the innate and adaptive immune system during infection by helminths can potentially uncover novel therapeutic applications against inflammatory, autoimmune and neoplastic diseases.
Collapse
Affiliation(s)
- Nathan M Ryan
- Department of Pathology, College of Medicine, Ohio State University Wexner Medical Center, Columbus, OH, U.S.A
| | - Steve Oghumu
- Department of Pathology, College of Medicine, Ohio State University Wexner Medical Center, Columbus, OH, U.S.A.
| |
Collapse
|
43
|
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: 4] [Impact Index Per Article: 0.7] [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
|
44
|
Buerfent BC, Gölz L, Hofmann A, Rühl H, Stamminger W, Fricker N, Hess T, Oldenburg J, Nöthen MM, Schumacher J, Hübner MP, Hoerauf A. Transcriptome-wide analysis of filarial extract-primed human monocytes reveal changes in LPS-induced PTX3 expression levels. Sci Rep 2019; 9:2562. [PMID: 30796272 PMCID: PMC6385373 DOI: 10.1038/s41598-019-38985-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 01/15/2019] [Indexed: 12/24/2022] Open
Abstract
Filarial nematodes modulate immune responses in their host to enable their survival and mediate protective effects against autoimmunity and allergies. In this study, we examined the immunomodulatory capacity of extracts from the human pathogenic filaria Brugia malayi (BmA) on human monocyte responses in a transcriptome-wide manner to identify associated pathways and diseases. As previous transcriptome studies often observed quiescent responses of innate cells to filariae, the potential of BmA to alter LPS driven responses was investigated by analyzing >47.000 transcripts of monocytes from healthy male volunteers stimulated with BmA, Escherichia coli LPS or a sequential stimulation of both. In comparison to ~2200 differentially expressed genes in LPS-only stimulated monocytes, only a limited number of differentially expressed genes were identified upon BmA priming before LPS re-stimulation with only PTX3↓ reaching statistical significance after correcting for multiple testing. Nominal significant differences were reached for metallothioneins↑, MMP9↑, CXCL5/ENA-78↑, CXCL6/GCP-2↑, TNFRSF21↓, and CCL20/MIP3α↓ and were confirmed by qPCR or ELISA. Flow cytometric analysis of activation markers revealed a reduced LPS-induced expression of HLA-DR and CD86 on BmA-primed monocytes as well as a reduced apoptosis of BmA-stimulated monocytes. While our experimental design does not allow a stringent extrapolation of our results to the development of filarial pathology, several genes that were identified in BmA-primed monocytes had previously been associated with filarial pathology, supporting the need for further research.
Collapse
Affiliation(s)
- B C Buerfent
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital of Bonn, Bonn, Germany
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
- Center for Human Genetics, University Hospital of Marburg, Marburg, Germany
| | - L Gölz
- Department of Orthodontics, Center of Dento-Maxillo-Facial Medicine, University of Bonn, Bonn, Germany
- Department of Orthodontics and Orofacial Orthopedics, University Hospital of Erlangen, Erlangen, Germany
| | - A Hofmann
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital of Bonn, Bonn, Germany
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | - H Rühl
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital of Bonn, Bonn, Germany
| | - W Stamminger
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital of Bonn, Bonn, Germany
| | - N Fricker
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | - T Hess
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
- Center for Human Genetics, University Hospital of Marburg, Marburg, Germany
| | - J Oldenburg
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital of Bonn, Bonn, Germany
| | - M M Nöthen
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | - J Schumacher
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
- Center for Human Genetics, University Hospital of Marburg, Marburg, Germany
| | - M P Hübner
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital of Bonn, Bonn, Germany.
| | - A Hoerauf
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital of Bonn, Bonn, Germany
- German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Bonn, Germany
| |
Collapse
|
45
|
Moyat M, Coakley G, Harris NL. The interplay of type 2 immunity, helminth infection and the microbiota in regulating metabolism. Clin Transl Immunology 2019; 8:e01089. [PMID: 31719981 PMCID: PMC6837856 DOI: 10.1002/cti2.1089] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 10/18/2019] [Accepted: 10/20/2019] [Indexed: 12/17/2022] Open
Abstract
Type 2 immunity has recently emerged as a critical player in metabolic status, with numerous studies investigating the role of type 2 immune cells within adipose tissue. Metabolic dysfunction is often characterised as a low-grade or chronic inflammatory state within tissues, and type 2 immunity may facilitate a return to metabolic homeostasis. A complex network of type 2 resident cells including M2 macrophages, eosinophils and ILC2s has been identified within adipose tissue. Although the effector cells in this equilibrium have not been clearly identified, any alteration of the type 2 microenvironment resulted in an altered metabolic state. Historically, the type 2 immune response has been associated with helminth infection. The type 2 immune response drives host resistance and plays an important role in promoting tissue repair following the migration of helminth larvae through tissues. Although helminths are largely eradicated in developed countries, infection rates remain high in poor communities within the developing world. Interestingly, there is strong evidence that helminth infection is inversely correlated with autoimmune or inflammatory disorders. Recently, an increasing amount of epidemiological and field studies suggest that it could be the same for obesity and metabolic syndrome. In the current review, we summarise the literature linking type 2 immunity to improved adipose tissue function. We then discuss more recent evidence indicating that helminth infection can provide protection against metabolic syndrome. Lastly, we explore the possible contributions of altered nutrient uptake, adipose tissue function and/or the intestinal microbiota with the ability of helminths to alter metabolic status.
Collapse
Affiliation(s)
- Mati Moyat
- Department of Immunology and PathologyMonash University Central Clinical SchoolMelbourneVICAustralia
| | - Gillian Coakley
- Department of Immunology and PathologyMonash University Central Clinical SchoolMelbourneVICAustralia
| | - Nicola L Harris
- Department of Immunology and PathologyMonash University Central Clinical SchoolMelbourneVICAustralia
| |
Collapse
|
46
|
Alexandre-Silva GM, Brito-Souza PA, Oliveira AC, Cerni FA, Zottich U, Pucca MB. The hygiene hypothesis at a glance: Early exposures, immune mechanism and novel therapies. Acta Trop 2018; 188:16-26. [PMID: 30165069 DOI: 10.1016/j.actatropica.2018.08.032] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 08/20/2018] [Accepted: 08/25/2018] [Indexed: 02/07/2023]
Abstract
The hygiene hypothesis was proposed almost three decades ago. Nevertheless, its mechanism still remains with relevant controversies. Some studies defend that early exposures during childhood to microbes and parasites are key determinants to prevent allergies and autoimmune diseases; however, other studies demonstrated that these early exposures can even potentiate the clinical scenario of the diseases. Based on several studies covering the influences of microbiome, parasites, related theories and others, this review focuses on recent advances in the hygiene hypothesis field. In addition, the main immunological mechanisms underlying the hygiene hypothesis are also discussed. We also strongly encourage that researchers do not consider the hygiene hypothesis as a theory based strictly on hygiene habits, but a theory combining diverse influences, as illustrated in this review as the hygiene hypothesis net.
Collapse
|
47
|
Sanya RE, Nkurunungi G, Andia Biraro I, Mpairwe H, Elliott AM. A life without worms. Trans R Soc Trop Med Hyg 2018; 111:3-11. [PMID: 28340138 PMCID: PMC5412073 DOI: 10.1093/trstmh/trx010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 02/27/2017] [Indexed: 12/14/2022] Open
Abstract
Worms have co-evolved with humans over millions of years. To survive, they manipulate host systems by modulating immune responses so that they cause (in the majority of hosts) relatively subtle harm. Anthelminthic treatment has been promoted as a measure for averting worm specific pathology and to mitigate subtle morbidities which may include effects on anaemia, growth, cognitive function and economic activity. With our changing environment marked by rapid population growth, urbanisation, better hygiene practices and anthelminthic treatment, there has been a decline in worm infections and other infectious diseases and a rise in non-communicable diseases such as allergy, diabetes and cardiovascular disease. This review reflects upon our age-old interaction with worms, and the broader ramifications of life without worms for vaccine responses and susceptibility to other infections, and for allergy-related and metabolic disease. We touch upon the controversy around the benefits of mass drug administration for the more-subtle morbidities that have been associated with worm infections and then focus our attention on broader, additional aspects of life without worms, which may be either beneficial or detrimental.
Collapse
Affiliation(s)
- Richard E Sanya
- MRC/UVRI Uganda Research Unit, Uganda Virus Research Institute, P.O. Box 49, Entebbe, Uganda.,College of Health Sciences, Makerere University, Kampala, Uganda
| | - Gyaviira Nkurunungi
- MRC/UVRI Uganda Research Unit, Uganda Virus Research Institute, P.O. Box 49, Entebbe, Uganda.,Department of Clinical Research, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | | | - Harriet Mpairwe
- MRC/UVRI Uganda Research Unit, Uganda Virus Research Institute, P.O. Box 49, Entebbe, Uganda
| | - Alison M Elliott
- MRC/UVRI Uganda Research Unit, Uganda Virus Research Institute, P.O. Box 49, Entebbe, Uganda.,Department of Clinical Research, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| |
Collapse
|
48
|
Tahapary DL, de Ruiter K, Martin I, Brienen EAT, van Lieshout L, Cobbaert CM, Soewondo P, Djuardi Y, Wiria AE, Houwing-Duistermaat JJ, Sartono E, Smit JWA, Yazdanbakhsh M, Supali T. Effect of Anthelmintic Treatment on Insulin Resistance: A Cluster-Randomized, Placebo-Controlled Trial in Indonesia. Clin Infect Dis 2018; 65:764-771. [PMID: 28472383 DOI: 10.1093/cid/cix416] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 04/28/2017] [Indexed: 01/14/2023] Open
Abstract
Background Emerging evidence suggests that helminth infections are associated with lower insulin resistance (IR). Current deworming programs might remove this helminth-associated protective effect. Therefore, we evaluated the anthelmintic treatment effect on changes in IR. Methods We conducted a double-blind, household-cluster-randomized, placebo-controlled clinical trial on Flores island, Indonesia, an area endemic for soil-transmitted helminths (STHs). All subjects received 4 rounds of albendazole or matching placebo with 3-month intervals, for 3 consecutive days. The primary outcome was the change in homeostatic model assessment of IR in those aged >16 years. An intention-to-treat analysis was performed involving all subjects and ad hoc in the helminth-infected subjects. Results We examined 797 (in 329 households) and 872 (in 353 households) subjects, who were assigned randomly into the albendazole and placebo arms, respectively. Albendazole was associated with a significant reduction in STH prevalence, total immunoglobulin E (IgE), and eosinophil count. Whereas albendazole had no effect on IR (estimated treatment effect, 0.006 [95% confidence interval, -.010 to .021]; P = .48) at the community level, it was associated with a significant increase in IR (estimated treatment effect, 0.031 [95% confidence interval, .004 to .059]; P = .04) (P value for interaction = .01) among helminth-infected subjects as detected by microscopy. Pathway analysis suggested that this might in part be due to an increased body mass index or a reduced eosinophil count. Conclusions Anthelmintic treatment reduces STH prevalence, total IgE, and eosinophil count but has no effect on IR at the community level. In helminth-infected subjects, treatment significantly increases IR, highlighting the need for metabolic health monitoring with ongoing deworming programs. Clinical Trials Registration ISRCTN 75636394.
Collapse
Affiliation(s)
- Dicky L Tahapary
- Department of Internal Medicine, Division of Endocrinology, Dr Cipto Mangunkusumo National General Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta.,Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Karin de Ruiter
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ivonne Martin
- Medical Statistics and Bioinformatics, Leiden University Medical Center, The Netherlands.,Department of Mathematics, Parahyangan Catholic University, Bandung, Indonesia
| | - Eric A T Brienen
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Lisette van Lieshout
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Christa M Cobbaert
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, The Netherlands
| | - Pradana Soewondo
- Department of Internal Medicine, Division of Endocrinology, Dr Cipto Mangunkusumo National General Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta
| | - Yenny Djuardi
- Department of Parasitology, Faculty of Medicine, Universitas Indonesia, Jakarta
| | - Aprilianto E Wiria
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands.,Department of Parasitology, Faculty of Medicine, Universitas Indonesia, Jakarta
| | - Jeanine J Houwing-Duistermaat
- Medical Statistics and Bioinformatics, Leiden University Medical Center, The Netherlands.,Department of Statistics, University of Leeds, United Kingdom
| | - Erliyani Sartono
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Johannes W A Smit
- Department of Internal Medicine, Radboud University Medical Centre, Nijmegen.,Department of Internal Medicine, Leiden University Medical Center, The Netherlands
| | - Maria Yazdanbakhsh
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Taniawati Supali
- Department of Parasitology, Faculty of Medicine, Universitas Indonesia, Jakarta
| |
Collapse
|
49
|
Bolus WR, Peterson KR, Hubler MJ, Kennedy AJ, Gruen ML, Hasty AH. Elevating adipose eosinophils in obese mice to physiologically normal levels does not rescue metabolic impairments. Mol Metab 2017; 8:86-95. [PMID: 29306658 PMCID: PMC5985044 DOI: 10.1016/j.molmet.2017.12.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 12/02/2017] [Accepted: 12/08/2017] [Indexed: 12/13/2022] Open
Abstract
Objective Obesity is a metabolic disorder that has reached epidemic proportions worldwide and leads to increased risk for diabetes, cardiovascular disease, asthma, certain cancers, and various other diseases. Obesity and its comorbidities are associated with impaired adipose tissue (AT) function. In the last decade, eosinophils have been identified as regulators of proper AT function. Our study aimed to determine whether normalizing the number of AT eosinophils in obese mice, to those of lean healthy mice, would reduce obesity and/or improve metabolic fitness. Methods C57BL/6J mice fed a high fat diet (HFD) were simultaneously given recombinant interleukin-5 (rIL5) for 8 weeks to increase AT eosinophils. Metabolic fitness was tested by evaluating weight gain, AT inflammation, glucose, lipid, and mixed-meal tolerance, AT insulin signaling, energy substrate utilization, energy expenditure, and white AT beiging capacity. Results Eosinophils were increased ∼3-fold in AT of obese HFD-fed mice treated with rIL5, and thus were restored to levels observed in lean healthy mice. However, there were no significant differences in rIL5-treated mice among the above listed comprehensive set of metabolic assays, despite the increased AT eosinophils. Conclusions We have shown that restoring obese AT eosinophils to lean healthy levels is not sufficient to allow for improvement in any of a range of metabolic features otherwise impaired in obesity. Thus, the mechanisms that identified eosinophils as positive regulators of AT function, and therefore systemic health, are more complex than initially understood and will require further study to fully elucidate. Adipose tissue eosinophils declined with high fat diet induced weight gain. Recombinant interleukin 5 treatment restored adipose eosinophils during obesity. Restoring adipose eosinophils didn't reduce weight gain or adipose mass. Restoring adipose eosinophils didn't rescue glucose tolerance or insulin signaling. Restoring adipose eosinophils didn't alter energy expenditure or beiging capacity.
Author Video Watch what authors say about their articles
Collapse
Affiliation(s)
- W Reid Bolus
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Kristin R Peterson
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Merla J Hubler
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Arion J Kennedy
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Marnie L Gruen
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Alyssa H Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA; VA Tennessee Valley Healthcare System, Nashville, TN, 37212, USA.
| |
Collapse
|
50
|
Aravindhan V, Anand G. Cell Type-Specific Immunomodulation Induced by Helminthes: Effect on Metainflammation, Insulin Resistance and Type-2 Diabetes. Am J Trop Med Hyg 2017; 97:1650-1661. [PMID: 29141759 DOI: 10.4269/ajtmh.17-0236] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Recent epidemiological studies have documented an inverse relationship between the decreasing prevalence of helminth infections and the increasing prevalence of metabolic diseases ("metabolic hygiene hypothesis"). Chronic inflammation leading to insulin resistance (IR) has now been identified as a major etiological factor for a variety of metabolic diseases other than obesity and Type-2 diabetes (metainflammation). One way by which helminth infections such as filariasis can modulate IR is by inducing a chronic, nonspecific, low-grade, immune suppression mediated by modified T-helper 2 (Th2) response (induction of both Th2 and regulatory T cells) which can in turn suppress the proinflammatory responses and promote insulin sensitivity (IS). This article provides evidence on how the cross talk between the innate and adaptive arms of the immune responses can modulate IR/sensitivity. The cross talk between innate (macrophages, dendritic cells, natural killer cells, natural killer T cells, myeloid derived suppressor cells, innate lymphoid cells, basophils, eosinophils, and neutrophils) and adaptive (helper T [CD4+] cells, cytotoxic T [CD8+] cells and B cells) immune cells forms two opposing circuits, one associated with IR and the other associated with IS under the conditions of metabolic syndrome and helminth-mediated immunomodulation, respectively.
Collapse
|