1
|
Mules TC, Vacca F, Cait A, Yumnam B, Schmidt A, Lavender B, Maclean K, Noble SL, Gasser O, Camberis M, Le Gros G, Inns S. A Small Intestinal Helminth Infection Alters Colonic Mucus and Shapes the Colonic Mucus Microbiome. Int J Mol Sci 2024; 25:12015. [PMID: 39596084 PMCID: PMC11593901 DOI: 10.3390/ijms252212015] [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: 09/13/2024] [Revised: 11/01/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
Infecting humans with controlled doses of small intestinal helminths, such as human hookworm, is proposed as a therapy for the colonic inflammatory disease ulcerative colitis. Strengthening the colonic mucus barrier is a potential mechanism by which small intestinal helminths could treat ulcerative colitis. In this study, we compare C57BL/6 mice infected with the small intestinal helminth Heligmosomoides polygyrus and uninfected controls to investigate changes in colonic mucus. Histology, gene expression, and immunofluorescent analysis demonstrate that this helminth induces goblet cell hyperplasia, and an upregulation of mucin sialylation, and goblet-cell-derived functional proteins resistin-like molecule-beta (RELM-β) and trefoil factors (TFFs), in the colon. Using IL-13 knockout mice, we reveal that these changes are predominantly IL-13-dependent. The assessment of the colonic mucus microbiome demonstrates that H. polygyrus infection increases the abundance of Ruminococcus gnavus, a commensal bacterium capable of utilising sialic acid as an energy source. This study also investigates a human cohort experimentally challenged with human hookworm. It demonstrates that TFF blood levels increase in individuals chronically infected with small intestinal helminths, highlighting a conserved mucus response between humans and mice. Overall, small intestinal helminths modify colonic mucus, highlighting this as a plausible mechanism by which human hookworm therapy could treat ulcerative colitis.
Collapse
Affiliation(s)
- Thomas C. Mules
- Malaghan Institute of Medical Research, Kelburn, Wellington 6012, New Zealand
- Department of Medicine, University of Otago, 23A Mein St., Newtown, Wellington 6242, New Zealand
| | - Francesco Vacca
- Malaghan Institute of Medical Research, Kelburn, Wellington 6012, New Zealand
| | - Alissa Cait
- Malaghan Institute of Medical Research, Kelburn, Wellington 6012, New Zealand
| | - Bibek Yumnam
- Malaghan Institute of Medical Research, Kelburn, Wellington 6012, New Zealand
| | - Alfonso Schmidt
- Hugh Green Technology Centre, Malaghan Institute of Medical Research, Wellington 6012, New Zealand
| | - Brittany Lavender
- Malaghan Institute of Medical Research, Kelburn, Wellington 6012, New Zealand
| | - Kate Maclean
- Malaghan Institute of Medical Research, Kelburn, Wellington 6012, New Zealand
| | - Sophia-Louise Noble
- Malaghan Institute of Medical Research, Kelburn, Wellington 6012, New Zealand
| | - Olivier Gasser
- Malaghan Institute of Medical Research, Kelburn, Wellington 6012, New Zealand
| | - Mali Camberis
- Malaghan Institute of Medical Research, Kelburn, Wellington 6012, New Zealand
| | - Graham Le Gros
- Malaghan Institute of Medical Research, Kelburn, Wellington 6012, New Zealand
| | - Stephen Inns
- Department of Medicine, University of Otago, 23A Mein St., Newtown, Wellington 6242, New Zealand
| |
Collapse
|
2
|
Xu X, Yuan J, Zhu M, Gao J, Meng X, Wu Y, Li X, Tong P, Chen H. The potential of orally exposed risk factors and constituents aggravating food allergy: Possible mechanism and target cells. Compr Rev Food Sci Food Saf 2024; 23:e70014. [PMID: 39230383 DOI: 10.1111/1541-4337.70014] [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: 04/11/2024] [Revised: 08/10/2024] [Accepted: 08/18/2024] [Indexed: 09/05/2024]
Abstract
Food allergy is a significant concern for the health of humans worldwide. In addition to dietary exposure of food allergens, genetic and environmental factors also play an important role in the development of food allergy. However, only the tip of the iceberg of risk factors in food allergy has been identified. The importance of food allergy caused by orally exposed risk factors and constituents, including veterinary drugs, pesticides, processed foods/derivatives, nanoparticles, microplastics, pathogens, toxins, food additives, dietary intake of salt/sugar/total fat, vitamin D, and therapeutic drugs, are highlighted and discussed in this review. Moreover, the epithelial barrier hypothesis, which is closely associated with the occurrence of food allergy, is also introduced. Additionally, several orally exposed risk factors and constituents that have been reported to disrupt the epithelial barrier are elucidated. Finally, the possible mechanisms and key immune cells of orally exposed risk factors and constituents in aggravating food allergy are overviewed. Further work should be conducted to define the specific mechanism by which these risk factors and constituents are driving food allergy, which will be of central importance to the targeted therapy of food allergy.
Collapse
Affiliation(s)
- Xiaoqian Xu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, P. R. China
- College of Food Science & Technology, Nanchang University, Nanchang, P. R. China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang, P. R. China
| | - Jin Yuan
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, P. R. China
- College of Food Science & Technology, Nanchang University, Nanchang, P. R. China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang, P. R. China
| | - Mengting Zhu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, P. R. China
- College of Food Science & Technology, Nanchang University, Nanchang, P. R. China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang, P. R. China
| | - Jinyan Gao
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, P. R. China
- College of Food Science & Technology, Nanchang University, Nanchang, P. R. China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang, P. R. China
| | - Xuanyi Meng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, P. R. China
- Sino-German Joint Research Institute, Nanchang University, Nanchang, P. R. China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang, P. R. China
| | - Yong Wu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, P. R. China
- Sino-German Joint Research Institute, Nanchang University, Nanchang, P. R. China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang, P. R. China
| | - Xin Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, P. R. China
- College of Food Science & Technology, Nanchang University, Nanchang, P. R. China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang, P. R. China
| | - Ping Tong
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, P. R. China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang, P. R. China
| | - Hongbing Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, P. R. China
- Sino-German Joint Research Institute, Nanchang University, Nanchang, P. R. China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang, P. R. China
| |
Collapse
|
3
|
Zhao YY, Wu ZJ, Hao SJ, Dong BB, Zheng YX, Liu B, Li J. Common alterations in parallel metabolomic profiling of serum and spinal cord and mechanistic studies on neuropathic pain following PPARα administration. Neuropharmacology 2024; 254:109988. [PMID: 38744401 DOI: 10.1016/j.neuropharm.2024.109988] [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: 09/22/2023] [Revised: 04/17/2024] [Accepted: 05/02/2024] [Indexed: 05/16/2024]
Abstract
Neuropathic pain (NP) is usually treated with analgesics and symptomatic therapy with poor efficacy and numerous side effects, highlighting the urgent need for effective treatment strategies. Recent studies have reported an important role for peroxisome proliferator-activated receptor alpha (PPARα) in regulating metabolism as well as inflammatory responses. Through pain behavioral assessment, we found that activation of PPARα prevented chronic constriction injury (CCI)-induced mechanical allodynia and thermal hyperalgesia. In addition, PPARα ameliorated inflammatory cell infiltration at the injury site and decreased microglial activation, NOD-like receptor protein 3 (NLRP3) inflammasome production, and spinal dendritic spine density, as well as improved serum and spinal cord metabolic levels in mice. Administration of PPARα antagonists eliminates the analgesic effect of PPARα agonists. PPARα relieves NP by inhibiting neuroinflammation and functional synaptic plasticity as well as modulating metabolic mechanisms, suggesting that PPARα may be a potential molecular target for NP alleviation. However, the effects of PPARα on neuroinflammation and synaptic plasticity should be further explored.
Collapse
Affiliation(s)
- Yu-Ying Zhao
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China
| | - Zi-Jun Wu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China
| | - Shu-Jing Hao
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China
| | - Bei-Bei Dong
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China
| | - Yu-Xin Zheng
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China
| | - Bin Liu
- Department of Critical Care Medicine, General Hospital of Tianjin Medical University, Tianjin, 300052, China; Center for Critical Care Medicine, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, 300020, China.
| | - Jing Li
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China.
| |
Collapse
|
4
|
Mules TC, Tang JS, Vacca F, Yumnam B, Schmidt A, Lavender B, Maclean K, Noble SL, Waugh C, van Ginkel R, Camberis M, Le Gros G, Inns S. Modulation of intestinal epithelial permeability by chronic small intestinal helminth infections. Immunol Cell Biol 2024; 102:396-406. [PMID: 38648862 DOI: 10.1111/imcb.12749] [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: 09/10/2023] [Revised: 03/18/2024] [Accepted: 03/28/2024] [Indexed: 04/25/2024]
Abstract
Increased permeability of the intestinal epithelial layer is linked to the pathogenesis and perpetuation of a wide range of intestinal and extra-intestinal diseases. Infecting humans with controlled doses of helminths, such as human hookworm (termed hookworm therapy), is proposed as a treatment for many of the same diseases. Helminths induce immunoregulatory changes in their host which could decrease epithelial permeability, which is highlighted as a potential mechanism through which helminths treat disease. Despite this, the influence of a chronic helminth infection on epithelial permeability remains unclear. This study uses the chronically infecting intestinal helminth Heligmosomoides polygyrus to reveal alterations in the expression of intestinal tight junction proteins and epithelial permeability during the infection course. In the acute infection phase (1 week postinfection), an increase in intestinal epithelial permeability is observed. Consistent with this finding, jejunal claudin-2 is upregulated and tricellulin is downregulated. By contrast, in the chronic infection phase (6 weeks postinfection), colonic claudin-1 is upregulated and epithelial permeability decreases. Importantly, this study also investigates changes in epithelial permeability in a small human cohort experimentally challenged with the human hookworm, Necator americanus. It demonstrates a trend toward small intestinal permeability increasing in the acute infection phase (8 weeks postinfection), and colonic and whole gut permeability decreasing in the chronic infection phase (24 weeks postinfection), suggesting a conserved epithelial response between humans and mice. In summary, our findings demonstrate dynamic changes in epithelial permeability during a chronic helminth infection and provide another plausible mechanism by which chronic helminth infections could be utilized to treat disease.
Collapse
Affiliation(s)
- Thomas C Mules
- Malaghan Institute of Medical Research, Wellington, New Zealand
- University of Otago, Wellington, New Zealand
| | - Jeffry S Tang
- Malaghan Institute of Medical Research, Wellington, New Zealand
- High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Francesco Vacca
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Bibek Yumnam
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Alfonso Schmidt
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | | | - Kate Maclean
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | | | | | | | - Mali Camberis
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Graham Le Gros
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Stephen Inns
- Malaghan Institute of Medical Research, Wellington, New Zealand
- University of Otago, Wellington, New Zealand
| |
Collapse
|
5
|
Vacca F, Mules TC, Camberis M, Lavender B, Noble SL, Cait A, Maclean K, Mamum J, Yumnam B, Te Kawa T, Ferrer-Font L, Tang JS, Gasser O, Le Gros G, Inns S. Controlled infection with cryopreserved human hookworm induces CTLA-4 expression on Tregs and upregulates tryptophan metabolism. Gut Microbes 2024; 16:2416517. [PMID: 39411786 PMCID: PMC11485773 DOI: 10.1080/19490976.2024.2416517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 09/11/2024] [Accepted: 10/04/2024] [Indexed: 10/19/2024] Open
Abstract
Infecting humans with controlled doses of helminths, such as human hookworm (termed hookworm therapy), is proposed to prevent or treat various intestinal and extraintestinal diseases. However, full-scale clinical trials examining hookworm therapy are limited by the inability to scale-up the production of hookworm larvae to infect sufficient numbers of patients. With the aim of overcoming this challenge, this study infected four healthy individuals with hookworm larvae that had been reanimated from cryopreserved eggs to examine their viability and immunogenicity. We demonstrate that reanimated cryopreserved hookworm larvae establish a viable hookworm infection and elicit a similar immune response to larvae cultured from fresh stool. Furthermore, a refined understanding of the therapeutic mechanisms of hookworm is imperative to determine which diseases to target with hookworm therapy. To investigate potential therapeutic mechanisms, this study assessed changes in the immune cells, microbiome, and plasma metabolome in the four healthy individuals infected with cryopreserved hookworm larvae and another nine individuals infected with larvae cultured from freshly obtained stool. We identified potential immunoregulatory mechanisms by which hookworm may provide a beneficial effect on its host, including increased expression of CTLA-4 on regulatory T cells (Tregs) and upregulation of tryptophan metabolism. Furthermore, we found that a participant's baseline microbiome predicted the severity of symptoms and intestinal inflammation experienced during a controlled hookworm infection. In summary, our findings demonstrate the feasibility of full-scale clinical trials examining hookworm therapy by minimizing the reliance on human donors and optimizing the culturing process, thereby enabling viable hookworm larvae to be mass-produced and enabling on-demand inoculation of patients. Furthermore, this study provides insights into the complex interactions between helminths and their host, which could inform the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Francesco Vacca
- Le Gros Laboratory, Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Thomas C. Mules
- Le Gros Laboratory, Malaghan Institute of Medical Research, Wellington, New Zealand
- Department of Medicine, University of Otago, Wellington, New Zealand
- Gastroenterology, Te Whatu Ora, Capital Coast and Hutt Valley, Wellington, New Zealand
| | - Mali Camberis
- Le Gros Laboratory, Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Brittany Lavender
- Le Gros Laboratory, Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Sophia-Louise Noble
- Le Gros Laboratory, Malaghan Institute of Medical Research, Wellington, New Zealand
- Department of Medicine, University of Otago, Wellington, New Zealand
| | - Alissa Cait
- Le Gros Laboratory, Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Kate Maclean
- Le Gros Laboratory, Malaghan Institute of Medical Research, Wellington, New Zealand
| | - John Mamum
- Le Gros Laboratory, Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Bibek Yumnam
- Le Gros Laboratory, Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Tama Te Kawa
- Le Gros Laboratory, Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Laura Ferrer-Font
- Hugh Green Technology Centre, Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Jeffry S. Tang
- Le Gros Laboratory, Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Olivier Gasser
- Le Gros Laboratory, Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Graham Le Gros
- Le Gros Laboratory, Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Stephen Inns
- Department of Medicine, University of Otago, Wellington, New Zealand
- Gastroenterology, Te Whatu Ora, Capital Coast and Hutt Valley, Wellington, New Zealand
| |
Collapse
|