1
|
Kumar H, Dhanjal DS, Dhalaria R, Kimta N, Cimler R, Kuča K. Dysbiosis significantly elevates the probability of altered affective function in Alzheimer disease (AD). INTERNATIONAL REVIEW OF NEUROBIOLOGY 2025; 180:1-24. [PMID: 40414630 DOI: 10.1016/bs.irn.2025.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2025]
Abstract
Changes in the makeup of gut microbiota are linked to many neuropsychiatric diseases. Although the exact connection between gut dysbiosis and brain dysfunction is not yet fully understood, but recent data suggests that gut dysbiosis may contribute to the development of Alzheimer's disease (AD) by promoting neuroinflammation, insulin resistance, oxidative stress, and amyloid-beta (Aβ) aggregation. Gut dysbiosis in animal models is primarily characterized by an elevated ratio of Firmicutes/Bacteroidetes which may lead to the accumulation of amyloid precursor protein (APP) in the intestine, in the early stages of AD. Probiotics play a significant role in preventing against the symptoms of AD by restoring gut-brain homeostasis. This chapter provides an overview of the gut microbiota and its dysregulation in etiology of AD. Moreover, novel insights into alteration of the composition of gut microbiota as a preventive or therapeutic approach to AD are discussed.
Collapse
Affiliation(s)
- Harsh Kumar
- Centre of Advanced Technologies, Faculty of Science, University of Hradec Kralove, Rokitanskeho, Hradec Kralove, Czech Republic
| | - Daljeet Singh Dhanjal
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India
| | - Rajni Dhalaria
- School of Biological and Environmental Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, India
| | - Neetika Kimta
- School of Biological and Environmental Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, India
| | - Richard Cimler
- Centre of Advanced Technologies, Faculty of Science, University of Hradec Kralove, Rokitanskeho, Hradec Kralove, Czech Republic
| | - Kamil Kuča
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic.
| |
Collapse
|
2
|
Dickson KB, Stadnyk AW, Zhou J, Lehmann C. Mucosal Immunity: Lessons from the Lower Respiratory and Small Intestinal Epithelia. Biomedicines 2025; 13:1052. [PMID: 40426880 PMCID: PMC12108762 DOI: 10.3390/biomedicines13051052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 05/29/2025] Open
Abstract
Mucosal epithelia represent a diverse group of tissues that function as a barrier against the external environment and exert a wide variety of tissue-specific secondary functions. This review focuses on the lower respiratory tract and small intestinal epithelia, which serve as two distinct sites within the body with respect to their physiological functions. This review provides an overview of their physiology, including both physiological and mechanical defense systems, and their immune responses, which allow both tissues to tolerate commensal organisms while mounting a response against potential pathogens. By highlighting the commonalities and differences across the two tissue types, opportunities to learn from these tissues emerge, which can inform the development of novel therapeutic strategies that harness the unique properties of mucosal epithelia.
Collapse
Affiliation(s)
- Kayle B. Dickson
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (K.B.D.); (A.W.S.)
| | - Andrew W. Stadnyk
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (K.B.D.); (A.W.S.)
- Department of Pediatrics, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Juan Zhou
- Department of Anesthesiology, Pain Management and Perioperative Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada;
| | - Christian Lehmann
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (K.B.D.); (A.W.S.)
- Department of Anesthesiology, Pain Management and Perioperative Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada;
- Department of Pharmacology, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
3
|
Zhang H, Su Q. Low-FODMAP Diet for Irritable Bowel Syndrome: Insights from Microbiome. Nutrients 2025; 17:544. [PMID: 39940404 PMCID: PMC11819959 DOI: 10.3390/nu17030544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 01/24/2025] [Accepted: 01/26/2025] [Indexed: 02/16/2025] Open
Abstract
Irritable bowel syndrome (IBS) is a prevalent gastrointestinal disorder characterized by chronic abdominal pain, bloating, and altered bowel habits. Low-FODMAP diets, which involve restricting fermentable oligosaccharides, disaccharides, monosaccharides, and polyols, have emerged as an effective dietary intervention for alleviating IBS symptoms. This review paper aims to synthesize current insights into the impact of a low-FODMAP diet on the gut microbiome and its mechanisms of action in managing IBS. We explore the alterations in microbial composition and function associated with a low-FODMAP diet and discuss the implications of these changes for gut health and symptom relief. Additionally, we examine the balance between symptom improvement and potential negative effects on microbial diversity and long-term gut health. Emerging evidence suggests that while a low-FODMAP diet can significantly reduce IBS symptoms, it may also lead to reductions in beneficial microbial populations. Strategies to mitigate these effects, such as the reintroduction phase and the use of probiotics, are evaluated. This review highlights the importance of a personalized approach to dietary management in IBS, considering individual variations in microbiome responses. Understanding the intricate relationship between diet, the gut microbiome, and IBS symptomatology will guide the development of more effective, sustainable dietary strategies for IBS patients.
Collapse
Affiliation(s)
- Haoshuai Zhang
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qi Su
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
4
|
Xu C, Aqib AI, Fatima M, Muneer S, Zaheer T, Peng S, Ibrahim EH, Li K. Deciphering the Potential of Probiotics in Vaccines. Vaccines (Basel) 2024; 12:711. [PMID: 39066349 PMCID: PMC11281421 DOI: 10.3390/vaccines12070711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 07/28/2024] Open
Abstract
The demand for vaccines, particularly those prepared from non-conventional sources, is rising due to the emergence of drug resistance around the globe. Probiotic-based vaccines are a wise example of such vaccines which represent new horizons in the field of vaccinology in providing an enhanced and diversified immune response. The justification for incorporating probiotics into vaccines lies in the fact that that they hold the capacity to regulate immune function directly or indirectly by influencing the gastrointestinal microbiota and related pathways. Several animal-model-based studies have also highlighted the efficacy of these vaccines. The aim of this review is to collect and summarize the trends in the recent scientific literature regarding the role of probiotics in vaccines and vaccinology, along with their impact on target populations.
Collapse
Affiliation(s)
- Chang Xu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Amjad Islam Aqib
- Department of Medicine, Cholistan University of Veterinary and Animal Sciences, Bahawalpur 63100, Pakistan
| | - Mahreen Fatima
- Faculty of Biosciences, Cholistan University of Veterinary and Animal Sciences, Bahawalpur 63100, Pakistan;
| | - Sadia Muneer
- Institute of Microbiology, University of Agriculture Faisalabad, Faisalabad 38000, Pakistan
| | - Tean Zaheer
- Department of Parasitology, University of Agriculture Faisalabad, Faisalabad 38000, Pakistan;
| | - Song Peng
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China;
| | - Essam H. Ibrahim
- Biology Department, Faculty of Science, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia
| | - Kun Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
5
|
Labetoulle M, Baudouin C, Benitez Del Castillo JM, Rolando M, Rescigno M, Messmer EM, Aragona P. How gut microbiota may impact ocular surface homeostasis and related disorders. Prog Retin Eye Res 2024; 100:101250. [PMID: 38460758 DOI: 10.1016/j.preteyeres.2024.101250] [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: 10/06/2023] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 03/11/2024]
Abstract
Changes in the bacterial flora in the gut, also described as gut microbiota, are readily acknowledged to be associated with several systemic diseases, especially those with an inflammatory, neuronal, psychological or hormonal factor involved in the pathogenesis and/or the perception of the disease. Maintaining ocular surface homeostasis is also based on all these four factors, and there is accumulating evidence in the literature on the relationship between gut microbiota and ocular surface diseases. The mechanisms involved are mostly interconnected due to the interaction of central and peripheral neuronal networks, inflammatory effectors and the hormonal system. A better understanding of the influence of the gut microbiota on the maintenance of ocular surface homeostasis, and on the onset or persistence of ocular surface disorders could bring new insights and help elucidate the epidemiology and pathology of ocular surface dynamics in health and disease. Revealing the exact nature of these associations could be of paramount importance for developing a holistic approach using highly promising new therapeutic strategies targeting ocular surface diseases.
Collapse
Affiliation(s)
- Marc Labetoulle
- Ophthalmology Départment, Hopital Bicetre, APHP, Université Paris-Saclay, IDMIT Infrastructure, Fontenay-aux-Roses Cedex, France; Hôpital National de la Vision des Quinze, Vingts, IHU ForeSight, Paris Saclay University, Paris, France.
| | - Christophe Baudouin
- Hôpital National de la Vision des Quinze, Vingts, IHU ForeSight, Paris Saclay University, Paris, France
| | - Jose M Benitez Del Castillo
- Departamento de Oftalmología, Hospital Clínico San Carlos, Clínica Rementeria, Instituto Investigaciones Oftalmologicas Ramon Castroviejo, Universidad Complutense, Madrid, Spain
| | - Maurizio Rolando
- Ocular Surface and Dry Eye Center, ISPRE Ophthalmics, Genoa, Italy
| | - Maria Rescigno
- IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, 20089, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, Pieve Emanuele, 20090, MI, Italy
| | | | - Pasquale Aragona
- Department of Biomedical Sciences, Ophthalmology Clinic, University of Messina, Messina, Italy
| |
Collapse
|
6
|
Liu Z, Zhang H, Lemaitre B, Li X. Duox activation in Drosophila Malpighian tubules stimulates intestinal epithelial renewal through a countercurrent flow. Cell Rep 2024; 43:114109. [PMID: 38613782 DOI: 10.1016/j.celrep.2024.114109] [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: 06/01/2021] [Revised: 02/26/2024] [Accepted: 03/28/2024] [Indexed: 04/15/2024] Open
Abstract
The gut must perform a dual role of protecting the host against toxins and pathogens while harboring mutualistic microbiota. Previous studies suggested that the NADPH oxidase Duox contributes to intestinal homeostasis in Drosophila by producing reactive oxygen species (ROS) in the gut that stimulate epithelial renewal. We find instead that the ROS generated by Duox in the Malpighian tubules leads to the production of Upd3, which enters the gut and stimulates stem cell proliferation. We describe in Drosophila the existence of a countercurrent flow system, which pushes tubule-derived Upd3 to the anterior part of the gut and stimulates epithelial renewal at a distance. Thus, our paper clarifies the role of Duox in gut homeostasis and describes the existence of retrograde fluid flow in the gut, collectively revealing a fascinating example of inter-organ communication.
Collapse
Affiliation(s)
- Zhonggeng Liu
- Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Hongyu Zhang
- Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Bruno Lemaitre
- Global Health Institute, School of Life Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| | - Xiaoxue Li
- Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, People's Republic of China; Global Health Institute, School of Life Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
7
|
da C. Pinaffi-Langley AC, Melia E, Hays FA. Exploring the Gut-Mitochondrial Axis: p66Shc Adapter Protein and Its Implications for Metabolic Disorders. Int J Mol Sci 2024; 25:3656. [PMID: 38612468 PMCID: PMC11011581 DOI: 10.3390/ijms25073656] [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: 02/17/2024] [Revised: 03/16/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
This review investigates the multifaceted role of the p66Shc adaptor protein and the gut microbiota in regulating mitochondrial function and oxidative stress, and their collective impact on the pathogenesis of chronic diseases. The study delves into the molecular mechanisms by which p66Shc influences cellular stress responses through Rac1 activation, Forkhead-type transcription factors inactivation, and mitochondria-mediated apoptosis, alongside modulatory effects of gut microbiota-derived metabolites and endotoxins. Employing an integrative approach, the review synthesizes findings from a broad array of studies, including molecular biology techniques and analyses of microbial metabolites' impacts on host cellular pathways. The results underscore a complex interplay between microbial metabolites, p66Shc activation, and mitochondrial dysfunction, highlighting the significance of the gut microbiome in influencing disease outcomes through oxidative stress pathways. Conclusively, the review posits that targeting the gut microbiota-p66Shc-mitochondrial axis could offer novel therapeutic strategies for mitigating the development and progression of metabolic diseases. This underscores the potential of dietary interventions and microbiota modulation in managing oxidative stress and inflammation, pivotal factors in chronic disease etiology.
Collapse
Affiliation(s)
- Ana Clara da C. Pinaffi-Langley
- Department of Nutritional Sciences, College of Allied Health, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA; (A.C.d.C.P.-L.); (E.M.)
| | - Elizabeth Melia
- Department of Nutritional Sciences, College of Allied Health, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA; (A.C.d.C.P.-L.); (E.M.)
| | - Franklin A. Hays
- Department of Nutritional Sciences, College of Allied Health, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA; (A.C.d.C.P.-L.); (E.M.)
- Stephenson Cancer Center, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA
| |
Collapse
|
8
|
Guo J, Li R, Ouyang Z, Tang J, Zhang W, Chen H, Zhu Q, Zhang J, Zhu G. Insights into the mechanism of transcription factors in Pb 2+-induced apoptosis. Toxicology 2024; 503:153760. [PMID: 38387706 DOI: 10.1016/j.tox.2024.153760] [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: 10/19/2023] [Revised: 02/12/2024] [Accepted: 02/19/2024] [Indexed: 02/24/2024]
Abstract
The health risks associated with exposure to heavy metals, such as Pb2+, are increasingly concerning the public. Pb2+ can cause significant harm to the human body through oxidative stress, autophagy, inflammation, and DNA damage, disrupting cellular homeostasis and ultimately leading to cell death. Among these mechanisms, apoptosis is considered crucial. It has been confirmed that transcription factors play a central role as mediators during the apoptosis process. Interestingly, these transcription factors have different effects on apoptosis depending on the concentration and duration of Pb2+ exposure. In this article, we systematically summarize the significant roles of several transcription factors in Pb2+-induced apoptosis. This information provides insights into therapeutic strategies and prognostic biomarkers for diseases related to Pb2+ exposure.
Collapse
Affiliation(s)
- Jingchong Guo
- The First Clinical Medical College of Nanchang University, Nanchang 330006, China
| | - Ruikang Li
- The First Clinical Medical College of Nanchang University, Nanchang 330006, China
| | - Zhuqing Ouyang
- The First Clinical Medical College of Nanchang University, Nanchang 330006, China
| | - Jiawen Tang
- The First Clinical Medical College of Nanchang University, Nanchang 330006, China
| | - Wei Zhang
- Department of Anatomy, Medical College of Nanchang University, Nanchang 330006, China
| | - Hui Chen
- Department of Anatomy, Medical College of Nanchang University, Nanchang 330006, China
| | - Qian Zhu
- Department of Anatomy, Medical College of Nanchang University, Nanchang 330006, China
| | - Jing Zhang
- Department of Anatomy, Medical College of Nanchang University, Nanchang 330006, China.
| | - Gaochun Zhu
- Department of Anatomy, Medical College of Nanchang University, Nanchang 330006, China.
| |
Collapse
|
9
|
Fernández Miyakawa ME, Casanova NA, Kogut MH. How did antibiotic growth promoters increase growth and feed efficiency in poultry? Poult Sci 2024; 103:103278. [PMID: 38052127 PMCID: PMC10746532 DOI: 10.1016/j.psj.2023.103278] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 11/04/2023] [Accepted: 11/12/2023] [Indexed: 12/07/2023] Open
Abstract
It has been hypothesized that reducing the bioenergetic costs of gut inflammation as an explanation for the effect of antibiotic growth promoters (AGPs) on animal efficiency, framing some observations but not explaining the increase in growth rate or the prevention of infectious diseases. The host's ability to adapt to alterations in environmental conditions and to maintain health involves managing all physiological interactions that regulate homeostasis. Thus, metabolic pathways are vital in regulating physiological health as the energetic demands of the host guides most biological functions. Mitochondria are not only the metabolic heart of the cell because of their role in energy metabolism and oxidative phosphorylation, but also a central hub of signal transduction pathways that receive messages about the health and nutritional states of cells and tissues. In response, mitochondria direct cellular and tissue physiological alterations throughout the host. The endosymbiotic theory suggests that mitochondria evolved from prokaryotes, emphasizing the idea that these organelles can be affected by some antibiotics. Indeed, therapeutic levels of several antibiotics can be toxic to mitochondria, but subtherapeutic levels may improve mitochondrial function and defense mechanisms by inducing an adaptive response of the cell, resulting in mitokine production which coordinates an array of adaptive responses of the host to the stressor(s). This adaptive stress response is also observed in several bacteria species, suggesting that this protective mechanism has been preserved during evolution. Concordantly, gut microbiome modulation by subinhibitory concentration of AGPs could be the result of direct stimulation rather than inhibition of determined microbial species. In eukaryotes, these adaptive responses of the mitochondria to internal and external environmental conditions, can promote growth rate of the organism as an evolutionary strategy to overcome potential negative conditions. We hypothesize that direct and indirect subtherapeutic AGP regulation of mitochondria functional output can regulate homeostatic control mechanisms in a manner similar to those involved with disease tolerance.
Collapse
Affiliation(s)
- Mariano Enrique Fernández Miyakawa
- Institute of Pathobiology, National Institute of Agricultural Technology (INTA), Argentina; National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina..
| | - Natalia Andrea Casanova
- Institute of Pathobiology, National Institute of Agricultural Technology (INTA), Argentina; National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - Michael H Kogut
- Southern Plains Agricultural Research Center, USDA-ARS, College Station, TX, USA
| |
Collapse
|
10
|
Zeng T, Fu Q, Luo F, Dai J, Fu R, Qi Y, Deng X, Lu Y, Xu Y. Lactic acid bacteria modulate the CncC pathway to enhance resistance to β-cypermethrin in the oriental fruit fly. THE ISME JOURNAL 2024; 18:wrae058. [PMID: 38618721 PMCID: PMC11069359 DOI: 10.1093/ismejo/wrae058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/08/2024] [Accepted: 04/08/2024] [Indexed: 04/16/2024]
Abstract
The gut microbiota of insects has been shown to regulate host detoxification enzymes. However, the potential regulatory mechanisms involved remain unknown. Here, we report that gut bacteria increase insecticide resistance by activating the cap "n" collar isoform-C (CncC) pathway through enzymatically generated reactive oxygen species (ROS) in Bactrocera dorsalis. We demonstrated that Enterococcus casseliflavus and Lactococcus lactis, two lactic acid-producing bacteria, increase the resistance of B. dorsalis to β-cypermethrin by regulating cytochrome P450 (P450) enzymes and α-glutathione S-transferase (GST) activities. These gut symbionts also induced the expression of CncC and muscle aponeurosis fibromatosis. BdCncC knockdown led to a decrease in resistance caused by gut bacteria. Ingestion of the ROS scavenger vitamin C in resistant strain affected the expression of BdCncC/BdKeap1/BdMafK, resulting in reduced P450 and GST activity. Furthermore, feeding with E. casseliflavus or L. lactis showed that BdNOX5 increased ROS production, and BdNOX5 knockdown affected the expression of the BdCncC/BdMafK pathway and detoxification genes. Moreover, lactic acid feeding activated the ROS-associated regulation of P450 and GST activity. Collectively, our findings indicate that symbiotic gut bacteria modulate intestinal detoxification pathways by affecting physiological biochemistry, thus providing new insights into the involvement of insect gut microbes in the development of insecticide resistance.
Collapse
Affiliation(s)
- Tian Zeng
- Guangdong Laboratory for Lingnan Modern Agriculture, Department of Entomology, South China Agricultural University, Guangzhou 510642, China
| | - Qianyan Fu
- Guangdong Laboratory for Lingnan Modern Agriculture, Department of Entomology, South China Agricultural University, Guangzhou 510642, China
| | - Fangyi Luo
- Guangdong Provincial Sericulture & Mulberry Engineering Research Center, Guangdong Prov Key Lab of AgroAnimal Genomics & Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Jian Dai
- Guangdong Laboratory for Lingnan Modern Agriculture, Department of Entomology, South China Agricultural University, Guangzhou 510642, China
| | - Rong Fu
- Guangdong Laboratory for Lingnan Modern Agriculture, Department of Entomology, South China Agricultural University, Guangzhou 510642, China
| | - Yixiang Qi
- Guangdong Laboratory for Lingnan Modern Agriculture, Department of Entomology, South China Agricultural University, Guangzhou 510642, China
| | - Xiaojuan Deng
- Guangdong Provincial Sericulture & Mulberry Engineering Research Center, Guangdong Prov Key Lab of AgroAnimal Genomics & Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Yongyue Lu
- Guangdong Laboratory for Lingnan Modern Agriculture, Department of Entomology, South China Agricultural University, Guangzhou 510642, China
| | - Yijuan Xu
- Guangdong Laboratory for Lingnan Modern Agriculture, Department of Entomology, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
11
|
Nolte S, Krüger K, Lenz C, Zentgraf K. Optimizing the Gut Microbiota for Individualized Performance Development in Elite Athletes. BIOLOGY 2023; 12:1491. [PMID: 38132317 PMCID: PMC10740793 DOI: 10.3390/biology12121491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/23/2023] [Accepted: 11/28/2023] [Indexed: 12/23/2023]
Abstract
The human gut microbiota can be compared to a fingerprint due to its uniqueness, hosting trillions of living organisms. Taking a sport-centric perspective, the gut microbiota might represent a physiological system that relates to health aspects as well as individualized performance in athletes. The athletes' physiology has adapted to their exceptional lifestyle over the years, including the diversity and taxonomy of the microbiota. The gut microbiota is influenced by several physiological parameters and requires a highly individual and complex approach to unravel the linkage between performance and the microbial community. This approach has been taken in this review, highlighting the functions that the microbial community performs in sports, naming gut-centered targets, and aiming for both a healthy and sustainable athlete and performance development. With this article, we try to consider whether initiating a microbiota analysis is practicable and could add value in elite sport, and what possibilities it holds when influenced through a variety of interventions. The aim is to support enabling a well-rounded and sustainable athlete and establish a new methodology in elite sport.
Collapse
Affiliation(s)
- Svenja Nolte
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, University of Giessen, 35394 Giessen, Germany; (K.K.); (C.L.)
| | - Karsten Krüger
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, University of Giessen, 35394 Giessen, Germany; (K.K.); (C.L.)
| | - Claudia Lenz
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, University of Giessen, 35394 Giessen, Germany; (K.K.); (C.L.)
| | - Karen Zentgraf
- Department 5: Psychology & Sports Sciences, Institute for Sports Sciences, Goethe University Frankfurt, 60323 Frankfurt am Main, Germany;
| |
Collapse
|
12
|
Mouradov D, Greenfield P, Li S, In EJ, Storey C, Sakthianandeswaren A, Georgeson P, Buchanan DD, Ward RL, Hawkins NJ, Skinner I, Jones IT, Gibbs P, Ma C, Liew YJ, Fung KYC, Sieber OM. Oncomicrobial Community Profiling Identifies Clinicomolecular and Prognostic Subtypes of Colorectal Cancer. Gastroenterology 2023; 165:104-120. [PMID: 36933623 DOI: 10.1053/j.gastro.2023.03.205] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 02/10/2023] [Accepted: 03/09/2023] [Indexed: 03/20/2023]
Abstract
BACKGROUND & AIMS Dysbiosis of gut microbiota is linked to the development of colorectal cancer (CRC). However, microbiota-based stratification of CRC tissue and how this relates to clinicomolecular characteristics and prognosis remains to be clarified. METHODS Tumor and normal mucosa from 423 patients with stage I to IV CRC were profiled by bacterial 16S rRNA gene sequencing. Tumors were characterized for microsatellite instability (MSI), CpG island methylator phenotype (CIMP), APC, BRAF, KRAS, PIK3CA, FBXW7, SMAD4, and TP53 mutations, subsets for chromosome instability (CIN), mutation signatures, and consensus molecular subtypes (CMS). Microbial clusters were validated in an independent cohort of 293 stage II/III tumors. RESULTS Tumors reproducibly stratified into 3 oncomicrobial community subtypes (OCSs) with distinguishing features: OCS1 (Fusobacterium/oral pathogens, proteolytic, 21%), right-sided, high-grade, MSI-high, CIMP-positive, CMS1, BRAF V600E, and FBXW7 mutated; OCS2 (Firmicutes/Bacteroidetes, saccharolytic, 44%), and OCS3 (Escherichia/Pseudescherichia/Shigella, fatty acid β-oxidation, 35%) both left-sided and exhibiting CIN. OCS1 was associated with MSI-related mutation signatures (SBS15, SBS20, ID2, and ID7) and OCS2 and OCS3 with SBS18 related to damage by reactive oxygen species. Among stage II/III patients, OCS1 and OCS3 both had poorer overall survival compared with OCS2 for microsatellite stable tumors (multivariate hazard ratio [HR], 1.85; 95% confidence interval [CI], 1.15-2.99; P = .012; and HR, 1.52; 95% CI 1.01-2.29; P = .044, respectively) and left-sided tumors (multivariate HR, 2.66; 95% CI, 1.45-4.86; P = .002; and HR, 1.76; 95% CI, 1.03-3.02; P = .039, respectively). CONCLUSIONS OCS classification stratified CRCs into 3 distinct subgroups with different clinicomolecular features and outcomes. Our findings provide a framework for a microbiota-based stratification of CRC to refine prognostication and to inform the development of microbiota-targeted interventions.
Collapse
Affiliation(s)
- Dmitri Mouradov
- Personalised Oncology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Paul Greenfield
- Energy Business Unit, Commonwealth Scientific and Industrial Research Organization, Lindfield, New South Wales, Australia; School of Natural Sciences, Macquarie University, North Ryde, New South Wales, Australia
| | - Shan Li
- Personalised Oncology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Eun-Jung In
- Personalised Oncology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Claire Storey
- Personalised Oncology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Anuratha Sakthianandeswaren
- Personalised Oncology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Peter Georgeson
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Melbourne, Victoria, Australia; University of Melbourne Center for Cancer Research, Victorian Comprehensive Cancer Center, Melbourne, Victoria, Australia
| | - Daniel D Buchanan
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Melbourne, Victoria, Australia; University of Melbourne Center for Cancer Research, Victorian Comprehensive Cancer Center, Melbourne, Victoria, Australia; Genomic Medicine and Family Cancer Clinic, The Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Robyn L Ward
- Prince of Wales Clinical School and Lowy Cancer Research Center, UNSW Sydney, Sydney, New South Wales, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Nicholas J Hawkins
- School of Medical Sciences, UNSW Sydney, Sydney, New South Wales, Australia
| | - Iain Skinner
- Department of Surgery, Western Health, Footscray, Victoria, Australia
| | - Ian T Jones
- Department of Surgery, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Peter Gibbs
- Personalised Oncology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia; Department of Medical Oncology, Western Health, St Albans, Victoria, Australia; Department of Medical Oncology, Western Health, Footscray, Victoria, Australia
| | - Chenkai Ma
- Molecular Diagnostics Solutions, Commonwealth Scientific and Industrial Research Organization Health and Biosecurity, Westmead, New South Wales, Australia
| | - Yi Jin Liew
- Molecular Diagnostics Solutions, Commonwealth Scientific and Industrial Research Organization Health and Biosecurity, Westmead, New South Wales, Australia
| | - Kim Y C Fung
- Molecular Diagnostics Solutions, Commonwealth Scientific and Industrial Research Organization Health and Biosecurity, Westmead, New South Wales, Australia
| | - Oliver M Sieber
- Personalised Oncology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia; Department of Surgery, The University of Melbourne, Parkville, Victoria, Australia; Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
13
|
Choudhury R, Gu Y, Bolhuis JE, Kleerebezem M. Early feeding leads to molecular maturation of the gut mucosal immune system in suckling piglets. Front Immunol 2023; 14:1208891. [PMID: 37304274 PMCID: PMC10248722 DOI: 10.3389/fimmu.2023.1208891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 05/16/2023] [Indexed: 06/13/2023] Open
Abstract
Introduction Diet-microbiota-host interactions are increasingly studied to comprehend their implications in host metabolism and overall health. Keeping in mind the importance of early life programming in shaping intestinal mucosal development, the pre-weaning period can be utilised to understand these interactions in suckling piglets. The objective of this study was to investigate the consequences of early life feeding on the time-resolved mucosal transcriptional program as well as mucosal morphology. Methods A customised fibrous feed was provided to piglets (early-fed or EF group; 7 litters) from five days of age until weaning (29 days of age) in addition to sow's milk, whereas control piglets (CON; 6 litters) suckled mother's milk only. Rectal swabs, intestinal content, and mucosal tissues (jejunum, colon) were obtained pre- and post-weaning for microbiota analysis (16S amplicon sequencing) and host transcriptome analysis (RNA sequencing). Results Early feeding accelerated both microbiota colonisation as well as host transcriptome, towards a more "mature state", with a more pronounced response in colon compared to jejunum. Early feeding elicited the largest impact on the colon transcriptome just before weaning (compared to post-weaning time-points), exemplified by the modulation of genes involved in cholesterol and energy metabolism and immune response. The transcriptional impact of early feeding persisted during the first days post-weaning and was highlighted by a stronger mucosal response to the weaning stress, via pronounced activation of barrier repair reactions, which is a combination of immune activation, epithelial migration and "wound-repair" like processes, compared to the CON piglets. Discussion Our study demonstrates the potential of early life nutrition in neonatal piglets as a means to support their intestinal development during the suckling period, and to improve adaptation during the weaning transition.
Collapse
Affiliation(s)
- Raka Choudhury
- Host-Microbe Interactomics Group, Department of Animal Sciences, Wageningen University & Research, Wageningen, Netherlands
| | - Yuner Gu
- Host-Microbe Interactomics Group, Department of Animal Sciences, Wageningen University & Research, Wageningen, Netherlands
| | - J. Elizabeth Bolhuis
- Adaptation Physiology Group, Department of Animal Sciences, Wageningen University & Research, Wageningen, Netherlands
| | - Michiel Kleerebezem
- Host-Microbe Interactomics Group, Department of Animal Sciences, Wageningen University & Research, Wageningen, Netherlands
| |
Collapse
|
14
|
Wei JD, Xu X. Oxidative stress in Wernicke's encephalopathy. Front Aging Neurosci 2023; 15:1150878. [PMID: 37261263 PMCID: PMC10229051 DOI: 10.3389/fnagi.2023.1150878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 05/02/2023] [Indexed: 06/02/2023] Open
Abstract
Wernicke's encephalopathy (WE) is a severe life-threatening disease that occurs due to vitamin B1 (thiamine) deficiency (TD). It is characterized by acute mental disorder, ataxia, and ophthalmoplegia. TD occurs because of the following reasons: insufficient intake, increased demand, and long-term drinking due to corresponding organ damage or failure. Recent studies showed that oxidative stress (OS) can damage organs and cause TD in the brain, which further leads to neurodegenerative diseases, such as WE. In this review, we discuss the effects of TD caused by OS on multiple organ systems, including the liver, intestines, and brain in WE. We believe that strengthening the human antioxidant system and reducing TD can effectively treat WE.
Collapse
Affiliation(s)
- Jun-Dong Wei
- Department of Basic Medical Science, Medical College, Taizhou University, Taizhou, China
| | - Xueming Xu
- Department of Psychiatry, Taizhou Second People's Hospital, Taizhou, China
| |
Collapse
|
15
|
Zhang X, Liu Y, Zhang T, Tan Y, Dai X, Yang YG, Zhang X. Advances in the potential roles of Cullin-RING ligases in regulating autoimmune diseases. Front Immunol 2023; 14:1125224. [PMID: 37006236 PMCID: PMC10064048 DOI: 10.3389/fimmu.2023.1125224] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/28/2023] [Indexed: 03/19/2023] Open
Abstract
Cullin-RING ligases (CRLs) are the largest class of E3 ubiquitin ligases regulating the stability and subsequent activity of a large number of important proteins responsible for the development and progression of various diseases, including autoimmune diseases (AIDs). However, the detailed mechanisms of the pathogenesis of AIDs are complicated and involve multiple signaling pathways. An in-depth understanding of the underlying regulatory mechanisms of the initiation and progression of AIDs will aid in the development of effective therapeutic strategies. CRLs play critical roles in regulating AIDs, partially by affecting the key inflammation-associated pathways such as NF-κB, JAK/STAT, and TGF-β. In this review, we summarize and discuss the potential roles of CRLs in the inflammatory signaling pathways and pathogenesis of AIDs. Furthermore, advances in the development of novel therapeutic strategies for AIDs through targeting CRLs are also highlighted.
Collapse
Affiliation(s)
- Xiaoying Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital, Jilin University, Changchun, China
| | - Yu’e Liu
- Tongji University Cancer Center, Shanghai Tenth People’s Hospital of Tongji University, School of Medicine, Tongji University, Shanghai, China
| | - Tong Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital, Jilin University, Changchun, China
| | - Yuying Tan
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital, Jilin University, Changchun, China
| | - Xiangpeng Dai
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital, Jilin University, Changchun, China
- *Correspondence: Xiangpeng Dai, ; Yong-Guang Yang, ; Xiaoling Zhang,
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital, Jilin University, Changchun, China
- International Center of Future Science, Jilin University, Changchun, China
- *Correspondence: Xiangpeng Dai, ; Yong-Guang Yang, ; Xiaoling Zhang,
| | - Xiaoling Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital, Jilin University, Changchun, China
- *Correspondence: Xiangpeng Dai, ; Yong-Guang Yang, ; Xiaoling Zhang,
| |
Collapse
|
16
|
Ramanathan R, Hatzios SK. Activity-based Tools for Interrogating Host Biology During Infection. Isr J Chem 2023; 63:e202200095. [PMID: 37744997 PMCID: PMC10512441 DOI: 10.1002/ijch.202200095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Indexed: 02/18/2023]
Abstract
Host cells sense and respond to pathogens by dynamically regulating cell signaling. The rapid modulation of signaling pathways is achieved by post-translational modifications (PTMs) that can alter protein structure, function, and/or binding interactions. By using chemical probes to broadly profile changes in enzyme function or side-chain reactivity, activity-based protein profiling (ABPP) can reveal PTMs that regulate host-microbe interactions. While ABPP has been widely utilized to uncover microbial mechanisms of pathogenesis, in this review, we focus on more recent applications of this technique to the discovery of host PTMs and enzymes that modulate signaling within infected cells. Collectively, these advances underscore the importance of ABPP as a tool for interrogating the host response to infection and identifying potential targets for host-directed therapies.
Collapse
Affiliation(s)
- Renuka Ramanathan
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT 06520 USA
- Microbial Sciences Institute, Yale University, West Haven, CT 06516 USA
| | - Stavroula K. Hatzios
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT 06520 USA
- Microbial Sciences Institute, Yale University, West Haven, CT 06516 USA
- Department of Chemistry, Yale University, New Haven, CT 06520 USA
| |
Collapse
|
17
|
A trans-kingdom T6SS effector induces the fragmentation of the mitochondrial network and activates innate immune receptor NLRX1 to promote infection. Nat Commun 2023; 14:871. [PMID: 36797302 PMCID: PMC9935632 DOI: 10.1038/s41467-023-36629-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/09/2023] [Indexed: 02/18/2023] Open
Abstract
Bacteria can inhibit the growth of other bacteria by injecting effectors using a type VI secretion system (T6SS). T6SS effectors can also be injected into eukaryotic cells to facilitate bacterial survival, often by targeting the cytoskeleton. Here, we show that the trans-kingdom antimicrobial T6SS effector VgrG4 from Klebsiella pneumoniae triggers the fragmentation of the mitochondrial network. VgrG4 colocalizes with the endoplasmic reticulum (ER) protein mitofusin 2. VgrG4 induces the transfer of Ca2+ from the ER to the mitochondria, activating Drp1 (a regulator of mitochondrial fission) thus leading to mitochondrial network fragmentation. Ca2+ elevation also induces the activation of the innate immunity receptor NLRX1 to produce reactive oxygen species (ROS). NLRX1-induced ROS limits NF-κB activation by modulating the degradation of the NF-κB inhibitor IκBα. The degradation of IκBα is triggered by the ubiquitin ligase SCFβ-TrCP, which requires the modification of the cullin-1 subunit by NEDD8. VgrG4 abrogates the NEDDylation of cullin-1 by inactivation of Ubc12, the NEDD8-conjugating enzyme. Our work provides an example of T6SS manipulation of eukaryotic cells via alteration of the mitochondria.
Collapse
|
18
|
Chen Z, Xiang Z, Cui L, Qin X, Chen S, Jin H, Zou H. Significantly different results in the ocular surface microbiome detected by tear paper and conjunctival swab. BMC Microbiol 2023; 23:31. [PMID: 36707800 PMCID: PMC9883858 DOI: 10.1186/s12866-023-02775-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/16/2023] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Great variation has been observed in the composition of the normal microbiota of the ocular surface, and therefore, in addition to differences in detection techniques, the method of collecting ocular surface specimens has a significant impact on the test results.The goal of this study is to ascertain whether the eye surface microbial communities detected by two different sampling methods are consistent and hence explore the feasibility of using tear test paper instead of conjunctival swabs to collect eye surface samples for microbial investigation. MATERIALS AND METHODS From July 15, 2021, to July 30, 2021, nonirritating tear test strips and conjunctival swabs of both eyes were used in 158 elderly people (> 60 years old) (79 diabetic and 79 nondiabetic adults) in Xinjing Community for high-throughput sequencing of the V3-V4 region of the 16S rRNA gene. The composition of the microbial communities in tear test paper and conjunctival swab samples was analyzed. RESULTS There was no statistically significant difference in Alpha diversity of ocular surface microorganisms represented by tear strip and conjunctival swab in diabetic group (P > 0.05), but there was statistically significant difference in Alpha diversity of ocular surface microorganisms detected by tear strip and conjunctival swab in nondiabetic group (P < 0.05). There were statistically significant differences in Beta diversity of ocular surface microorganisms detected by two sampling methods between diabetic group and nondiabetic group (P < 0.05). There were statistically significant differences in ocular surface microorganisms detected by tear strip method between diabetic group and nondiabetic group (P < 0.05), but there was no statistically significant difference in conjunctival swab method (P > 0.05). CONCLUSIONS Tear test paper and conjunctival swabs detect different compositions of microbes through two different techniques of eye surface microbe sampling. Tear test paper cannot completely replace conjunctival swab specimens for the study of microbes related to eye surface diseases.
Collapse
Affiliation(s)
- Zhangling Chen
- grid.16821.3c0000 0004 0368 8293Department of Ophthalmology, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (Preparatory Stage), Shanghai, China ,grid.412478.c0000 0004 1760 4628Department of Ophthalmology, Shanghai General Hospital, Nanjing Medical University, Hongkou District, No. 100, Haining Road, Shanghai, 200080 China
| | - Zhaoyu Xiang
- grid.16821.3c0000 0004 0368 8293Department of Ophthalmology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lipu Cui
- grid.16821.3c0000 0004 0368 8293Department of Ophthalmology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xinran Qin
- grid.16821.3c0000 0004 0368 8293Department of Ophthalmology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shuli Chen
- grid.16821.3c0000 0004 0368 8293Department of Ophthalmology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Huiyi Jin
- grid.16821.3c0000 0004 0368 8293Department of Ophthalmology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Haidong Zou
- grid.412478.c0000 0004 1760 4628Department of Ophthalmology, Shanghai General Hospital, Nanjing Medical University, Hongkou District, No. 100, Haining Road, Shanghai, 200080 China ,grid.16821.3c0000 0004 0368 8293Department of Ophthalmology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China ,grid.412478.c0000 0004 1760 4628Shanghai Key Laboratory of Fundus Diseases, Shanghai, China ,Shanghai Eye Diseases Prevention & Treatment Center/Shanghai Eye Hospital, Shanghai, China ,grid.412478.c0000 0004 1760 4628National Clinical Research Center for Eye Diseases, Shanghai, China ,grid.412478.c0000 0004 1760 4628Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| |
Collapse
|
19
|
Li D, Xu Y, Wang M, Fang S, Li SH, Cui Y. Differences of semen microbiota among breeding boars with different reproductive ages. J Anim Sci 2023; 101:skad247. [PMID: 37478469 PMCID: PMC10424712 DOI: 10.1093/jas/skad247] [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: 04/07/2023] [Accepted: 07/21/2023] [Indexed: 07/23/2023] Open
Abstract
In this study, we investigated 18 healthy and fertile Duroc boars, dividing them into two groups based on their reproductive age: 9 boars aged 18 mo and 9 boars aged 36 mo. Prior to semen sampling, all boars were raised together under identical management conditions for a period of 3 mo. Our findings revealed that older boars exhibited lower sperm motility and a higher proportion of abnormal sperm morphology compared to younger boars. Furthermore, older boars demonstrated lower anti-oxidant capacity in their semen, as indicated by elevated levels of malondialdehyde and decreased levels of superoxide dismutase and glutathione peroxidase. Microbiota analysis utilizing the 16S rRNA technique showed that the semen microbiota of older boars had reduced alpha-diversity and beta-diversity in comparison to younger boars. We identified the Streptococcus genus and Streptococcus gallolyticus subsp macedonicus species served as biomarkers for semen from younger breeding boars, while the Bacteroides pyogenes species as a biomarker for semen from older breeding boars. Additionally, the semen from older boars exhibited a higher abundance of Aerococcus, Gallicola, Ulvibacter, and Proteiniphilum compared to younger boars. Spearman correlation analysis showed that these four bacteria were negatively correlated with semen quality. The abundance of Gallicola and Proteiniphilum were negatively correlated with semen anti-oxidant capacity. Additionally, the reduction of semen anti-oxidant capacity was correlated to the decrease of semen quality. Based on these findings, we concluded that the semen of older boars contains a higher abundance of harmful bacteria, which contributes to the observed reduction in semen anti-oxidant capacity and overall semen quality in this group.
Collapse
Affiliation(s)
- Desheng Li
- College of Animal Science and Veterinary Medicine, Jinzhou Medical University, Jinzhou 121001, China
| | - Yunhe Xu
- College of Animal Science and Veterinary Medicine, Jinzhou Medical University, Jinzhou 121001, China
| | - Mi Wang
- College of Animal Science and Veterinary Medicine, Jinzhou Medical University, Jinzhou 121001, China
| | - Shan Fang
- College of Animal Science and Veterinary Medicine, Jinzhou Medical University, Jinzhou 121001, China
| | - Shi Han Li
- College of Animal Science and Veterinary Medicine, Jinzhou Medical University, Jinzhou 121001, China
| | - Yan Cui
- College of Animal Science and Veterinary Medicine, Jinzhou Medical University, Jinzhou 121001, China
| |
Collapse
|
20
|
Ahmed LA, Al-Massri KF. Gut Microbiota Modulation for Therapeutic Management of Various Diseases: A New Perspective Using Stem Cell Therapy. Curr Mol Pharmacol 2023; 16:43-59. [PMID: 35196976 DOI: 10.2174/1874467215666220222105004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 11/08/2021] [Accepted: 12/16/2021] [Indexed: 11/22/2022]
Abstract
Dysbiosis has been linked to various diseases ranging from cardiovascular, neurologic, gastrointestinal, respiratory, and metabolic illnesses to cancer. Restoring of gut microbiota balance represents an outstanding clinical target for the management of various multidrug-resistant diseases. Preservation of gut microbial diversity and composition could also improve stem cell therapy which now has diverse clinical applications in the field of regenerative medicine. Gut microbiota modulation and stem cell therapy may be considered a highly promising field that could add up towards the improvement of different diseases, increasing the outcome and efficacy of each other through mutual interplay or interaction between both therapies. Importantly, more investigations are required to reveal the cross-talk between microbiota modulation and stem cell therapy to pave the way for the development of new therapies with enhanced therapeutic outcomes. This review provides an overview of dysbiosis in various diseases and their management. It also discusses microbiota modulation via antibiotics, probiotics, prebiotics, and fecal microbiota transplant to introduce the concept of dysbiosis correction for the management of various diseases. Furthermore, we demonstrate the beneficial interactions between microbiota modulation and stem cell therapy as a way for the development of new therapies in addition to limitations and future challenges regarding the applications of these therapies.
Collapse
Affiliation(s)
- Lamiaa A Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Khaled F Al-Massri
- Department of Pharmacy and Biotechnology, Faculty of Medicine and Health Sciences, University of Palestine, Gaza, Palestine
| |
Collapse
|
21
|
Das TK, Ganesh BP. Interlink between the gut microbiota and inflammation in the context of oxidative stress in Alzheimer's disease progression. Gut Microbes 2023; 15:2206504. [PMID: 37127846 PMCID: PMC10153019 DOI: 10.1080/19490976.2023.2206504] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/03/2023] Open
Abstract
The microbiota-gut-brain axis is an important pathway of communication and may dynamically contribute to Alzheimer's disease (AD) pathogenesis. Pathological commensal gut microbiota alterations, termed as dysbiosis, can influence intestinal permeability and break the blood-brain barrier which may trigger AD pathogenesis via redox signaling, neuronal, immune, and metabolic pathways. Dysbiosis increases the oxidative stress. Oxidants affect the innate immune system through recognizing microbial-derived pathogens by Toll-like receptors and initiating the inflammatory process. Most of the gut microbiome research work highlights the relationship between the gut microbiota and AD, but the contributory connection between precise bacteria and brain dysfunction in AD pathology cannot be fully demonstrated. Here, we summarize the current information of the fundamental connections between oxidative stress, inflammation, and gut dysbiosis in AD. This review emphasizes on the involvement of gut microbiota in the regulation of oxidative stress, inflammation, immune responses including central and peripheral cross-talk. It provides insights for novel preventative and therapeutic approaches in AD.
Collapse
Affiliation(s)
- Tushar K Das
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Bhanu P Ganesh
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
22
|
Chen Z, Jia Y, Xiao Y, Lin Q, Qian Y, Xiang Z, Cui L, Qin X, Chen S, Yang C, Zou H. Microbiological Characteristics of Ocular Surface Associated With Dry Eye in Children and Adolescents With Diabetes Mellitus. Invest Ophthalmol Vis Sci 2022; 63:20. [PMID: 36538004 PMCID: PMC9769747 DOI: 10.1167/iovs.63.13.20] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Purpose To analyze the characteristics of ocular surface microbial composition in children and adolescents with diabetes mellitus and dry eye (DE) by tear analysis. Methods We selected 65 children and adolescents aged 8 to 16 years with DE and non-DE diabetes mellitus and 33 healthy children in the same age group from the Shanghai Children and Adolescent Diabetes Eye Study. Tears were collected for high-throughput sequencing of the V3 and V4 region of 16S rRNA. The ocular surface microbiota in diabetic DE (DM-DE; n = 31), diabetic with non-DE (DM-NDE; n = 34), and healthy (NDM; n = 33) groups were studied. QIIME2 software was used to analyze the microbiota of each group. Results The DM-DE group had the highest amplicon sequence variants, and the differences in α-diversity and β-diversity of micro-organisms in the ocular surfaces of DM-DE, diabetic with non-DE, and healthy eyes were statistically significant (P < 0.05). Bacteroidetes (15.6%), Tenericutes (9.3%), Firmicutes (21.8%), and Lactococcus (7.9%), Bacteroides (7.8%), Acinetobacter (3.9%), Clostridium (0.8%), Lactobacillus (0.8%) and Streptococcus (0.2%) were the specific phyla and genera, respectively, in the DM-DE group. Conclusions Compared with the patients with non-DE and healthy children, the microbial diversity of the ocular surface in children and adolescents with diabetes mellitus and DE was higher with unique bacterial phyla and genera composition.
Collapse
Affiliation(s)
- Zhangling Chen
- Department of Ophthalmology, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (Preparatory Stage), Shanghai, China,Department of Ophthalmology, Shanghai General Hospital, Nanjing Medical University, Shanghai, China
| | - Yan Jia
- Department of Ophthalmology, Children's Hospital of Fudan University, Shanghai, China
| | - Ying Xiao
- Department of Ophthalmology, Children's Hospital of Fudan University, Shanghai, China
| | - Qiurong Lin
- Shanghai Eye Diseases Prevention & Treatment Center/Shanghai Eye Hospital, Shanghai, China
| | - Yu Qian
- Department of Ophthalmology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhaoyu Xiang
- Department of Ophthalmology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lipu Cui
- Department of Ophthalmology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xinran Qin
- Department of Ophthalmology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shuli Chen
- Department of Ophthalmology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chenhao Yang
- Department of Ophthalmology, Children's Hospital of Fudan University, Shanghai, China
| | - Haidong Zou
- Department of Ophthalmology, Shanghai General Hospital, Nanjing Medical University, Shanghai, China,Shanghai Eye Diseases Prevention & Treatment Center/Shanghai Eye Hospital, Shanghai, China,Department of Ophthalmology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China,Shanghai Key Laboratory of Fundus Diseases, Shanghai, China,National Clinical Research Center for Eye Diseases, Shanghai, China,Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| |
Collapse
|
23
|
Ma N, Chen X, Johnston LJ, Ma X. Gut microbiota-stem cell niche crosstalk: A new territory for maintaining intestinal homeostasis. IMETA 2022; 1:e54. [PMID: 38867904 PMCID: PMC10989768 DOI: 10.1002/imt2.54] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 08/04/2022] [Accepted: 08/14/2022] [Indexed: 06/14/2024]
Abstract
Intestinal epithelium undergoes rapid cellular turnover, relying on the local niche, to support intestinal stem cells (ISCs) function and self-renewal. Research into the association between ISCs and disease continues to expand at a rapid rate. However, the detailed interaction of ISCs and gut microbes remains to be elucidated. Thus, this review witnessed major advances in the crosstalk between ISCs and gut microbes, delivering key insights into (1) construction of ISC niche and molecular mechanism of how to jointly govern epithelial homeostasis and protect against intestinal diseases with the participation of Wnt, bone morphogenetic protein, and Notch; (2) differentiation fate of ISCs affect the gut microbiota. Meanwhile, the presence of intestinal microbes also regulates ISC function; (3) microbiota regulation on ISCs by Wnt and Notch signals through pattern recognition receptors; (4) how do specific microbiota-related postbiotics influence ISCs to maintain intestinal epithelial regeneration and homeostasis that provide insights into a promising alternative therapeutic method for intestinal diseases. Considering the detailed interaction is still unclear, it is necessary to further explore the regulatory role of gut microbiota on ISCs to utilize microbes to alleviate gut disorders. Furthermore, these major advances collectively drive us ever closer to breakthroughs in regenerative medicine and cancer treatment by microbial transplantation in the clinic.
Collapse
Affiliation(s)
- Ning Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and TechnologyChina Agricultural UniversityBeijingChina
| | - Xiyue Chen
- State Key Laboratory of Animal Nutrition, College of Animal Science and TechnologyChina Agricultural UniversityBeijingChina
| | - Lee J. Johnston
- West Central Research & Outreach CenterUniversity of MinnesotaMorrisMinnesotaUSA
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and TechnologyChina Agricultural UniversityBeijingChina
| |
Collapse
|
24
|
Julien C, Anakok E, Treton X, Nachury M, Nancey S, Buisson A, Fumery M, Filippi J, Maggiori L, Panis Y, Zerbib P, François Y, Dubois A, Sabbagh C, Rahili A, Seksik P, Allez M, Lefevre JH, Le Corff S, Bonnet A, Beyer-Berjot L, Sokol H. Impact of the Ileal Microbiota on Surgical Site Infections in Crohn's Disease: A Nationwide Prospective Cohort. J Crohns Colitis 2022; 16:1211-1221. [PMID: 35218661 DOI: 10.1093/ecco-jcc/jjac026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/24/2021] [Accepted: 02/25/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Surgery is performed in 50-70% of Crohn's disease [CD] patients, and its main risk is surgical site infection [SSI]. The microbiota has been extensively assessed in CD but not as a potential risk factor for septic morbidity. The objective of this study was to assess the impact of the gut microbiota on SSI in CD. METHODS We used the multicentric REMIND prospective cohort to identify all patients who experienced SSI after ileocolonic resection for CD, defined as any postoperative local septic complication within 90 days after surgery: wound abscess, intra-abdominal collection, anastomotic leakage or enterocutaneous fistula. The mucosa-associated microbiota of the ileal resection specimen was analysed by 16S gene sequencing in 149 patients. The variable selection and prediction were performed with random forests [R package VSURF] on clinical and microbiotal data. The criterion of performance that we considered was the area under the Receiver Operating Characteristic [ROC] curve [AUC]. RESULTS SSI occurred in 24 patients [16.1%], including 15 patients [10.1%] with major morbidity. There were no significant differences between patients with or without SSI regarding alpha and beta diversity. The top selected variables for the prediction of SSI were all microbiota-related. The maximum AUC [0.796] was obtained with a model including 14 genera, but an AUC of 0.78 had already been obtained with a model including only six genera [Hungatella, Epulopiscium, Fusobacterium, Ruminococcaceae_ucg_009, Actinomyces and Ralstonia]. CONCLUSION The gut microbiota has the potential to predict SSI after ileocolonic resection for CD. It might play a role in this frequent postoperative complication.
Collapse
Affiliation(s)
- Clément Julien
- Department of Gastrointestinal Surgery, Hôpital Nord, Assistance Publique - Hôpitaux de Marseille, Aix-Marseille Univ., Chemin des Bourrely, 13015 Marseille, France
| | - Emré Anakok
- Sorbonne Université, UMR CNRS 8001, LPSM, 75005 Paris, France.,Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Gastroenterology Department, F-75012 Paris, France
| | - Xavier Treton
- Gastroenterology Department Hôpital Beaujon, MICI et Assistance Nutritive, Clichy, France
| | - Maria Nachury
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Stéphane Nancey
- Gastroenterology Department, Lyon Sud Hospital, Hospices Civils de Lyon, and INSERM U1111, CIRI, Lyon, France
| | - Anthony Buisson
- Gastroenterology Department, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Mathurin Fumery
- Hepatogastroenterology Department, Amiens University Hospital, Amiens, France
| | - Jérôme Filippi
- Gastroenterology Department, Hopital Archet 2, Nice, France
| | - Léon Maggiori
- Digestive, Oncologic, and Endocrine Surgery Department, Hôpital Saint-Louis, AP-HP, Université de Paris, Paris, France
| | - Yves Panis
- Department of Colorectal Surgery, Beaujon Hospital and University of Paris, France
| | - Philippe Zerbib
- Digestive Surgery and Transplantation, Claude Huriez Hospital, CHRU de Lille, Lille Université Nord de France, Lille, France
| | - Yves François
- Surgery Department, Lyon Sud Hospital, Hospices Civils de Lyon , Lyon, France
| | - Anne Dubois
- Surgery Department, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Charles Sabbagh
- Surgery Department, Amiens University Hospital, Amiens, France
| | - Amine Rahili
- Surgery Department, Hopital Archet 2, Nice, France
| | - Philippe Seksik
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Gastroenterology Department, F-75012 Paris, France.,Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Matthieu Allez
- Gastroenterology Department, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Jérémie H Lefevre
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France.,Sorbonne Université, Department of Digestive Surgery, AP-HP, Hôpital Saint Antoine, F-75012, Paris, France
| | | | - Sylvain Le Corff
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France.,Samovar, Télécom SudParis, Institut Polytechnique de Paris , Paris, France
| | - Anna Bonnet
- Sorbonne Université, UMR CNRS 8001, LPSM, 75005 Paris, France.,Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Laura Beyer-Berjot
- Department of Gastrointestinal Surgery, Hôpital Nord, Assistance Publique - Hôpitaux de Marseille, Aix-Marseille Univ., Chemin des Bourrely, 13015 Marseille, France.,Laboratoire de biomécanique appliquée (LBA), UMR T24, Aix-Marseille Univ/Université Gustave Eiffel, Boulevard Pierre Dramard, Marseille, France.,Centre for Surgical Teaching and Research (CERC), Aix-Marseille Univ, Boulevard Pierre Dramard, Marseille, France
| | - Harry Sokol
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Gastroenterology Department, F-75012 Paris, France.,Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France.,INRA, UMR1319 Micalis & AgroParisTech, Jouy en Josas, France
| |
Collapse
|
25
|
Ezzine C, Loison L, Montbrion N, Bôle-Feysot C, Déchelotte P, Coëffier M, Ribet D. Fatty acids produced by the gut microbiota dampen host inflammatory responses by modulating intestinal SUMOylation. Gut Microbes 2022; 14:2108280. [PMID: 35978476 PMCID: PMC9466625 DOI: 10.1080/19490976.2022.2108280] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The gut microbiota produces a wide variety of metabolites, which interact with intestinal cells and contribute to host physiology. The effect of gut commensal bacteria on host protein SUMOylation, an essential ubiquitin-like modification involved in various intestinal functions, remains, however, unknown. Here, we show that short chain fatty acids (SCFAs) and branched chain fatty acids (BCFAs) produced by the gut microbiota increase protein SUMOylation in intestinal cells in a pH-dependent manner. We demonstrate that these metabolites inactivate intestinal deSUMOylases and promote the hyperSUMOylation of nuclear matrix-associated proteins. We further show that BCFAs inhibit the NF-κB pathway, decrease pro-inflammatory cytokine expression, and promote intestinal epithelial integrity. Together, our results reveal that fatty acids produced by gut commensal bacteria regulate intestinal physiology by modulating SUMOylation and illustrate a new mechanism of dampening of host inflammatory responses triggered by the gut microbiota.
Collapse
Affiliation(s)
- Chaima Ezzine
- Université de Rouen Normandie, INSERM UMR 1073, Nutrition, Inflammation et axe Microbiote-Intestin-Cerveau, Rouen, France,Université de Rouen Normandie, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Léa Loison
- Université de Rouen Normandie, INSERM UMR 1073, Nutrition, Inflammation et axe Microbiote-Intestin-Cerveau, Rouen, France,Université de Rouen Normandie, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Nadine Montbrion
- Université de Rouen Normandie, INSERM UMR 1073, Nutrition, Inflammation et axe Microbiote-Intestin-Cerveau, Rouen, France,Université de Rouen Normandie, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Christine Bôle-Feysot
- Université de Rouen Normandie, INSERM UMR 1073, Nutrition, Inflammation et axe Microbiote-Intestin-Cerveau, Rouen, France,Université de Rouen Normandie, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Pierre Déchelotte
- Université de Rouen Normandie, INSERM UMR 1073, Nutrition, Inflammation et axe Microbiote-Intestin-Cerveau, Rouen, France,Université de Rouen Normandie, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France,Department of Nutrition, Rouen University Hospital, CHU Rouen, Rouen, France
| | - Moïse Coëffier
- Université de Rouen Normandie, INSERM UMR 1073, Nutrition, Inflammation et axe Microbiote-Intestin-Cerveau, Rouen, France,Université de Rouen Normandie, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France,Department of Nutrition, Rouen University Hospital, CHU Rouen, Rouen, France
| | - David Ribet
- Université de Rouen Normandie, INSERM UMR 1073, Nutrition, Inflammation et axe Microbiote-Intestin-Cerveau, Rouen, France,Université de Rouen Normandie, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France,CONTACT David Ribet Université de Rouen Normandie, INSERM UMR 1073, Nutrition, Inflammation et axe Microbiote-Intestin-Cerveau, UFR Santé - 22 Boulevard Gambetta, 76000Rouen, France; Université de Rouen Normandie, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| |
Collapse
|
26
|
Massai L, Messori L, Carpentieri A, Amoresano A, Melchiorre C, Fiaschi T, Modesti A, Gamberi T, Magherini F. The effects of two gold-N-heterocyclic carbene (NHC) complexes in ovarian cancer cells: a redox proteomic study. Cancer Chemother Pharmacol 2022; 89:809-823. [PMID: 35543764 PMCID: PMC9135895 DOI: 10.1007/s00280-022-04438-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/15/2022] [Indexed: 01/04/2023]
Abstract
PURPOSE Ovarian cancer is the fifth leading cause of cancer-related deaths in women. Standard treatment consists of tumor debulking surgery followed by platinum and paclitaxel chemotherapy; yet, despite the initial response, about 70-75% of patients develop resistance to chemotherapy. Gold compounds represent a family of very promising anticancer drugs. Among them, we previously investigated the cytotoxic and pro-apoptotic properties of Au(NHC) and Au(NHC)2PF6, i.e., a monocarbene gold(I) complex and the corresponding bis(carbene) complex. Gold compounds are known to alter the redox state of cells interacting with free cysteine and selenocysteine residues of several proteins. Herein, a redox proteomic study has been carried out to elucidate the mechanisms of cytotoxicity in A2780 human ovarian cancer cells. METHODS A biotinylated iodoacetamide labeling method coupled with mass spectrometry was used to identify oxidation-sensitive protein cysteines. RESULTS Gold carbene complexes cause extensive oxidation of several cellular proteins; many affected proteins belong to two major functional classes: carbohydrate metabolism, and cytoskeleton organization/cell adhesion. Among the affected proteins, Glyceraldehyde-3-phosphate dehydrogenase inhibition was proved by enzymatic assays and by ESI-MS studies. We also found that Au(NHC)2PF6 inhibits mitochondrial respiration impairing complex I function. Concerning the oxidized cytoskeletal proteins, gold binding to the free cysteines of actin was demonstrated by ESI-MS analysis. Notably, both gold compounds affected cell migration and invasion. CONCLUSIONS In this study, we deepened the mode of action of Au(NHC) and Au(NHC)2PF6, identifying common cellular targets but confirming their different influence on the mitochondrial function.
Collapse
Affiliation(s)
- Lara Massai
- Department of Chemistry 'Ugo Schiff', University of Florence, via della Lastruccia 3-13, Sesto Fiorentino, 50019, Firenze, Italy
| | - Luigi Messori
- Department of Chemistry 'Ugo Schiff', University of Florence, via della Lastruccia 3-13, Sesto Fiorentino, 50019, Firenze, Italy
| | - Andrea Carpentieri
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
| | - Angela Amoresano
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
| | - Chiara Melchiorre
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
| | - Tania Fiaschi
- Department of Experimental and Clinical Biomedical Sciences, Mario Serio" University of Florence Viale G.B. Morgagni 50, 50134, Florence, Italy
| | - Alessandra Modesti
- Department of Experimental and Clinical Biomedical Sciences, Mario Serio" University of Florence Viale G.B. Morgagni 50, 50134, Florence, Italy
| | - Tania Gamberi
- Department of Experimental and Clinical Biomedical Sciences, Mario Serio" University of Florence Viale G.B. Morgagni 50, 50134, Florence, Italy.
| | - Francesca Magherini
- Department of Experimental and Clinical Biomedical Sciences, Mario Serio" University of Florence Viale G.B. Morgagni 50, 50134, Florence, Italy.
| |
Collapse
|
27
|
Kociszewska D, Vlajkovic SM. The Association of Inflammatory Gut Diseases with Neuroinflammatory and Auditory Disorders. Front Biosci (Elite Ed) 2022; 14:8. [PMID: 35730449 DOI: 10.31083/j.fbe1402008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/11/2022] [Accepted: 02/24/2022] [Indexed: 11/06/2022]
Abstract
Disorders such as inflammatory bowel disease (IBD) and celiac disease (CeD) result in intestinal hyperpermeability or 'leaky' gut. The increased permeability of the intestinal barrier allows microbial metabolites, toxins, and pathogens to infiltrate the bloodstream and extraintestinal tissues, causing systemic inflammation. Despite differences in aetiology and pathophysiology, IBD and CeD share several extraintestinal manifestations such as neuroinflammation, neurological and psychiatric manifestations, and sensorineural hearing loss (SNHL). This narrative review focuses on the association between intestinal hyperpermeability with the brain and inner ear diseases. We postulate that the microbial metabolites and pathogens released from the gut increase the permeability of natural barriers, such as the blood-brain barrier (BBB) and blood-labyrinth barrier (BLB). The barrier breakdown allows the spreading of inflammatory processes to the brain and inner ear, leading to disease.
Collapse
Affiliation(s)
- Dagmara Kociszewska
- Department of Physiology and The Eisdell Moore Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, 1142 Auckland, New Zealand
| | - Srdjan M Vlajkovic
- Department of Physiology and The Eisdell Moore Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, 1142 Auckland, New Zealand
| |
Collapse
|
28
|
Wang G, Yuan J, Luo J, Ocansey DKW, Zhang X, Qian H, Xu W, Mao F. Emerging role of protein modification in inflammatory bowel disease. J Zhejiang Univ Sci B 2022; 23:173-188. [PMID: 35261214 PMCID: PMC8913920 DOI: 10.1631/jzus.b2100114] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 08/17/2021] [Indexed: 11/11/2022]
Abstract
The onset of inflammatory bowel disease (IBD) involves many factors, including environmental parameters, microorganisms, and the immune system. Although research on IBD continues to expand, the specific pathogenesis mechanism is still unclear. Protein modification refers to chemical modification after protein biosynthesis, also known as post-translational modification (PTM), which causes changes in the properties and functions of proteins. Since proteins can be modified in different ways, such as acetylation, methylation, and phosphorylation, the functions of proteins in different modified states will also be different. Transitions between different states of protein or changes in modification sites can regulate protein properties and functions. Such modifications like neddylation, sumoylation, glycosylation, and acetylation can activate or inhibit various signaling pathways (e.g., nuclear factor-κB (NF-κB), extracellular signal-regulated kinase (ERK), and protein kinase B (AKT)) by changing the intestinal flora, regulating immune cells, modulating the release of cytokines such as interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), and interferon-γ (IFN-γ), and ultimately leading to the maintenance of the stability of the intestinal epithelial barrier. In this review, we focus on the current understanding of PTM and describe its regulatory role in the pathogenesis of IBD.
Collapse
Affiliation(s)
- Gaoying Wang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, China
- Clinical Laboratory, Wuxi Maternal and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi 214002, China
| | - Jintao Yuan
- Clinical Laboratory, the People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Zhenjiang 212300, China
| | - Ji Luo
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Dickson Kofi Wiredu Ocansey
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, China
- Directorate of University Health Services, University of Cape Coast, Cape Coast 02630, Ghana
| | - Xu Zhang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Hui Qian
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Wenrong Xu
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, China.
| |
Collapse
|
29
|
Koskimäki JJ, Pohjanen J, Kvist J, Fester T, Härtig C, Podolich O, Fluch S, Edesi J, Häggman H, Pirttilä AM. The meristem-associated endosymbiont Methylorubrum extorquens DSM13060 reprograms development and stress responses of pine seedlings. TREE PHYSIOLOGY 2022; 42:391-410. [PMID: 34328183 PMCID: PMC8842435 DOI: 10.1093/treephys/tpab102] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 07/19/2021] [Indexed: 06/13/2023]
Abstract
Microbes living in plant tissues-endophytes-are mainly studied in crop plants where they typically colonize the root apoplast. Trees-a large carbon source with a high capacity for photosynthesis-provide a variety of niches for endophytic colonization. We have earlier identified a new type of plant-endophyte interaction in buds of adult Scots pine, where Methylorubrum species live inside the meristematic cells. The endosymbiont Methylorubrum extorquens DSM13060 significantly increases needle and root growth of pine seedlings without producing plant hormones, but by aggregating around host nuclei. Here, we studied gene expression and metabolites of the pine host induced by M. extorquens DSM13060 infection. Malic acid was produced by pine to potentially boost M. extorquens colonization and interaction. Based on gene expression, the endosymbiont activated the auxin- and ethylene (ET)-associated hormonal pathways through induction of CUL1 and HYL1, and suppressed salicylic and abscisic acid signaling of pine. Infection by the endosymbiont had an effect on pine meristem and leaf development through activation of GLP1-7 and ALE2, and suppressed flowering, root hair and lateral root formation by downregulation of AGL8, plantacyanin, GASA7, COW1 and RALFL34. Despite of systemic infection of pine seedlings by the endosymbiont, the pine genes CUL1, ETR2, ERF3, HYL, GLP1-7 and CYP71 were highly expressed in the shoot apical meristem, rarely in needles and not in stem or root tissues. Low expression of MERI5, CLH2, EULS3 and high quantities of ononitol suggest that endosymbiont promotes viability and protects pine seedlings against abiotic stress. Our results indicate that the endosymbiont positively affects host development and stress tolerance through mechanisms previously unknown for endophytic bacteria, manipulation of plant hormone signaling pathways, downregulation of senescence and cell death-associated genes and induction of ononitol biosynthesis.
Collapse
Affiliation(s)
- Janne J Koskimäki
- Ecology and Genetics Research Unit, University of Oulu, Paavo Havaksentie J1, FI-90014 Oulu, Finland
| | - Johanna Pohjanen
- Ecology and Genetics Research Unit, University of Oulu, Paavo Havaksentie J1, FI-90014 Oulu, Finland
| | - Jouni Kvist
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, FI-00014 Helsinki, Finland
| | - Thomas Fester
- Department of Environmental Microbiology, Helmholtz Centre for Environmental Research – UFZ, Permoserstr. 15, 04318 Leipzig, Germany
| | - Claus Härtig
- Department of Environmental Microbiology, Helmholtz Centre for Environmental Research – UFZ, Permoserstr. 15, 04318 Leipzig, Germany
| | - Olga Podolich
- Institute of Molecular Biology and Genetics of NASU, Acad. Zabolotnoho str., 150 03680 Kyiv, Ukraine
| | | | - Jaanika Edesi
- Ecology and Genetics Research Unit, University of Oulu, Paavo Havaksentie J1, FI-90014 Oulu, Finland
- Production Systems, Tree Breeding, Natural Resources Institute Finland LUKE, FI-57200 Savonlinna, Finland
| | - Hely Häggman
- Ecology and Genetics Research Unit, University of Oulu, Paavo Havaksentie J1, FI-90014 Oulu, Finland
| | | |
Collapse
|
30
|
Watanabe D, Kamada N. Contribution of the Gut Microbiota to Intestinal Fibrosis in Crohn's Disease. Front Med (Lausanne) 2022; 9:826240. [PMID: 35198577 PMCID: PMC8859331 DOI: 10.3389/fmed.2022.826240] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/13/2022] [Indexed: 12/16/2022] Open
Abstract
In Crohn's disease (CD), intestinal fibrosis is a critical determinant of a patient's prognosis. Although inflammation may be a prerequisite for the initiation of intestinal fibrosis, research shows that the progression or continuation of intestinal fibrosis can occur independently of inflammation. Thus, once initiated, intestinal fibrosis may persist even if medical treatment controls inflammation. Clearly, an understanding of the pathophysiological mechanisms of intestinal fibrosis is required to diminish its occurrence. Accumulating evidence suggests that the gut microbiota contributes to the pathogenesis of intestinal fibrosis. For example, the presence of antibodies against gut microbes can predict which CD patients will have intestinal complications. In addition, microbial ligands can activate intestinal fibroblasts, thereby inducing the production of extracellular matrix. Moreover, in various animal models, bacterial infection can lead to the development of intestinal fibrosis. In this review, we summarize the current knowledge of the link between intestinal fibrosis in CD and the gut microbiota. We highlight basic science and clinical evidence that the gut microbiota can be causative for intestinal fibrosis in CD and provide valuable information about the animal models used to investigate intestinal fibrosis.
Collapse
Affiliation(s)
- Daisuke Watanabe
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Nobuhiko Kamada
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
- WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| |
Collapse
|
31
|
Zhong M, Sun A, Xiao T, Yao G, Sang L, Zheng X, Zhang J, Jin X, Xu L, Yang W, Wang P, Hu K, Zhang D, Ge J. A Randomized, Single-Blind, Group Sequential, Active-Controlled Study to Evaluate the Clinical Efficacy and Safety of α-Lipoic Acid for Critically Ill Patients With Coronavirus Disease 2019 (COVID-19). Front Med (Lausanne) 2022; 8:566609. [PMID: 35186959 PMCID: PMC8854372 DOI: 10.3389/fmed.2021.566609] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 11/15/2021] [Indexed: 12/15/2022] Open
Abstract
OBJECT To evaluate the clinical efficacy and safety of α-Lipoic acid (ALA) for critically ill patients with coronavirus disease 2019 (COVID-19). METHODS A randomized, single-blind, group sequential, active-controlled trial was performed at JinYinTan Hospital, Wuhan, China. Between February 2020 and March 2020, 17 patients with critically ill COVID-19 were enrolled in our study. Eligible patients were randomly assigned in a 1:1 ratio to receive either ALA (1200 mg/d, intravenous infusion) once daily plus standard care or standard care plus equal volume saline infusion (placebo) for 7 days. All patients were monitored within the 7 days therapy and followed up to day 30 after therapy. The primary outcome of this study was the Sequential Organ Failure Estimate (SOFA) score, and the secondary outcome was the all-cause mortality within 30 days. RESULT Nine patients were randomized to placebo group and 8 patients were randomized to ALA group. SOFA score was similar at baseline, increased from 4.3 to 6.0 in the placebo group and increased from 3.8 to 4.0 in the ALA group (P = 0.36) after 7 days. The 30-day all-cause mortality tended to be lower in the ALA group (3/8, 37.5%) compared to that in the placebo group (7/9, 77.8%, P = 0.09). CONCLUSION In our study, ALA use is associated with lower SOFA score increase and lower 30-day all-cause mortality as compared with the placebo group. Although the mortality rate was two-folds higher in placebo group than in ALA group, only borderline statistical difference was evidenced due to the limited patient number. Future studies with larger patient cohort are warranted to validate the role of ALA in critically ill patients with COVID-19. CLINICAL TRIAL REGISTRATION http://www.chictr.org.cn/showproj.aspx?proj=49534.
Collapse
Affiliation(s)
- Ming Zhong
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Wuhan JinYinTan Hospital, Wuhan, China
| | - Aijun Sun
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
- Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Ting Xiao
- Intensive Care Unit of the Six Floor of South Building, Wuhan JinYinTan Hospital, Wuhan, China
| | - Ge Yao
- Intensive Care Unit of the Six Floor of South Building, Wuhan JinYinTan Hospital, Wuhan, China
| | - Ling Sang
- Department of Critical Care Medicine, GuangZhou Institute of Respiratory Health, The First Affiliated Hospital of GuangZhou Medical University, Guangzhou, China
| | - Xia Zheng
- Department of Critical Care Medicine, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Jinyan Zhang
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
| | - Xuejuan Jin
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
| | - Lei Xu
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
| | - Wenlong Yang
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
| | - Peng Wang
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
| | - Kai Hu
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
| | | | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
- Institute of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
32
|
Waziri A, Bharti C, Aslam M, Jamil P, Mirza MA, Javed MN, Pottoo U, Ahmadi A, Alam MS. Probiotics for the Chemoprotective Role against the Toxic Effect of Cancer Chemotherapy. Anticancer Agents Med Chem 2022; 22:654-667. [PMID: 33992067 DOI: 10.2174/1871520621666210514000615] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/10/2020] [Accepted: 01/05/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Chemo- and radiation therapy-based clinical management of different types of cancers is associated with toxicity and several side effects. Therefore, there is always an unmet need to explore agents that reduce such risk factors. Among these, natural products have attracted much attention because of their potent antioxidant and antitumor effects. In the past, some breakthrough outcomes established that various bacteria in the human intestinal gut are bearing growth-promoting attributes and suppressing the conversion of pro-carcinogens into carcinogens. Hence probiotics integrated approaches are nowadays being explored as rationalized therapeutics in the clinical management of cancer. METHODS Here, published literature was explored to review chemoprotective roles of probiotics against toxic and side effects of chemotherapeutics. RESULTS Apart from excellent anti-cancer abilities, probiotics alleviate toxicity & side effects of chemotherapeutics, with a high degree of safety and efficiency. CONCLUSION Preclinical and clinical evidence suggests that due to the chemoprotective roles of probiotics against side effects and toxicity of chemotherapeutics, their integration in chemotherapy would be a judicious approach.
Collapse
Affiliation(s)
- Aafrin Waziri
- University School of Biotechnology, Guru Gobind Singh Indraprastha University, Delhi, India
| | - Charu Bharti
- School of Medical and Allied Sciences, K.R. Mangalam University, Sohna Road, Gurgaon, Haryana-122103, India
| | - Mohammed Aslam
- Faculty of Pharmacy, AL Hawash Private University, Homs, Syria
| | - Parween Jamil
- Faculty of Dentistry, Jamia Millia Islamia, New Delhi, India
| | - Mohd Aamir Mirza
- Department of Pharmaceutics, Faculty of Pharmacy, Jamia Hamdard, New Delhi, India
| | - Md Noushad Javed
- Department of Pharmacy, SMAS, KR Mangalam University, Gurugram, India
- Department of Pharmaceutics, Faculty of Pharmacy, Jamia Hamdard, New Delhi, India
| | - Uzma Pottoo
- Department of Food Science & Technology, School of Applied Sciences & Technology, University of Kashmir, J.K., India
| | - Amirhossein Ahmadi
- Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Md Sabir Alam
- NIMS Institute of Pharmacy, NIMS University, NH-11C, Delhi - Jaipur Expy, Shobha Nagar, Jaipur, Rajasthan India
- SGT College of Pharmacy, SGT University, Gurgaon-Badli Road Chandu, Budhera, Gurugram, Haryana 122505, India
| |
Collapse
|
33
|
Dey P, Chaudhuri SR, Efferth T, Pal S. The intestinal 3M (microbiota, metabolism, metabolome) zeitgeist - from fundamentals to future challenges. Free Radic Biol Med 2021; 176:265-285. [PMID: 34610364 DOI: 10.1016/j.freeradbiomed.2021.09.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/18/2021] [Accepted: 09/30/2021] [Indexed: 12/12/2022]
Abstract
The role of the intestine in human health and disease has historically been neglected and was mostly attributed to digestive and absorptive functions. In the past two decades, however, discoveries related to human nutrition and intestinal host-microbe reciprocal interaction have established the essential role of intestinal health in the pathogenesis of chronic diseases and the overall wellbeing. That transfer of gut microbiota could be a means of disease phenotype transfer has revolutionized our understanding of chronic disease pathogenesis. This narrative review highlights the major concepts related to intestinal microbiota, metabolism, and metabolome (3M) that have facilitated our fundamental understanding of the association between the intestine, and human health and disease. In line with increased interest of microbiota-dependent modulation of human health by dietary phytochemicals, we have also discussed the emerging concepts beyond the phytochemical bioactivities which emphasizes the integral role of microbial metabolites of parent phytochemicals at extraintestinal tissues. Finally, this review concludes with challenges and future prospects in defining the 3M interactions and has emphasized the fact that, it takes 'guts' to stay healthy.
Collapse
Affiliation(s)
- Priyankar Dey
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, Punjab, India.
| | - Saumya Ray Chaudhuri
- Council of Scientific and Industrial Research (CSIR), Institute of Microbial Technology, Chandigarh, India
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany
| | - Sirshendu Pal
- Mukherjee Hospital, Mitra's Clinic and Nursing Home, Siliguri, West Bengal, India
| |
Collapse
|
34
|
Salman HA, Yaakop AS, Al-Mustafa A, Tarawneh K, Aladaileh S, Al-Rimawi F, Alakhras F, Abu-Lafi S, Zarzour RA, Wahab H. The dual impact of Jordanian Ephedra alte for inhibiting pepsin and treating microbial infections. Saudi J Biol Sci 2021; 28:6245-6253. [PMID: 34764751 PMCID: PMC8568995 DOI: 10.1016/j.sjbs.2021.06.090] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 06/26/2021] [Accepted: 06/27/2021] [Indexed: 11/24/2022] Open
Abstract
Screening of phytochemical Ephedra alte crude extract by GC–MS and HPLC analysis indicated the presence of alkaloids, tannins, flavonoids, terpenoids, and phenolic acid in the extract. The total phenolic content of E. alte methanol extract was 39.43 mg of Gallic acid eq/g, crude E. alte with 56.74, and 2.42 µg Trolox equivalent antioxidant capacity (TEAC)/g of plant extract according to DPPH and FRAP assay, respectively. The antimicrobial activity of E. alte against Staphylococcus aureus, staphylococcus epidermidis, Escherichia coli, and Klebsiellaoxytoca demonstrated a mean zone diameter of inhibition ranging from 0 to 17 mm. The MIC of the extracts ranged from 0.5 to 1.0 mg/mL. E. alte extract inhibits pepsin enzyme activity with IC50 values of 213.67 µg/ml. This study revealed that E. alte extract has pepsin enzyme inhibitory, antibacterial, antioxidant activities. The current outcomes indicate that E. alte might be employed as a natural agent for managing GERD and infectious diseases.
Collapse
Affiliation(s)
- Haya Ayyal Salman
- School of Biological Sciences, Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Amira Suriaty Yaakop
- School of Biological Sciences, Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Ahmed Al-Mustafa
- Department of Biological Sciences, Faculty of Science, Mutah University, P.O. Box 7, Mutah 61710, Al-Karak, Jordan
| | - Khaled Tarawneh
- Department of Biological Sciences, Faculty of Science, Mutah University, P.O. Box 7, Mutah 61710, Al-Karak, Jordan
| | - Saleem Aladaileh
- Department of Pharmacy Practice, College of Pharmacy, University of Hafr Al-Batin, Hafr Al-Batin, Saudi Arabia
| | - Fuad Al-Rimawi
- Chemistry Department, Faculty of Science and Technology, Al-Quds University, P.O. Box 2002, Jerusalem, Palestine
| | - Fadi Alakhras
- Department of Chemistry, College of Science, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | - Saleh Abu-Lafi
- Faculty of Pharmacy, Al-Quds University, P.O. Box 20002, Jerusalem, Palestine
| | | | - Habibah Wahab
- School of Pharmaceutical Sciences, University Sains Malaysia, Malaysia
| |
Collapse
|
35
|
Stanojević S, Blagojević V, Ćuruvija I, Vujić V. Lactobacillus rhamnosus Affects Rat Peritoneal Cavity Cell Response to Stimulation with Gut Microbiota: Focus on the Host Innate Immunity. Inflammation 2021; 44:2429-2447. [PMID: 34505975 DOI: 10.1007/s10753-021-01513-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 07/05/2021] [Indexed: 12/01/2022]
Abstract
Gut microbiota contribute to shaping the immune repertoire of the host, whereas probiotics may exert beneficial effects by modulating immune responses. Having in mind the differences in both the composition of gut microbiota and the immune response between rats of Albino Oxford (AO) and Dark Agouti (DA) rat strains, we investigated if intraperitoneal (i.p.) injection of live Lactobacillus rhamnosus (LB) may influence peritoneal cavity cell response to in vitro treatments with selected microbiota in the rat strain-dependent manner. Peritoneal cavity cells from AO and DA rats were lavaged two (d2) and seven days (d7) following i.p. injection with LB and tested for NO, urea, and H2O2 release basally, or upon in vitro stimulation with autologous E.coli and Enterococcus spp. Whereas the single i.p. injection of LB nearly depleted resident macrophages and increased the proportion of small inflammatory macrophages and monocytes on d2 in both rat strains, greater proportion of MHCIIhiCD163- and CCR7+ cells and increased NO/diminished H2O2 release in DA compared with AO rats suggest a more intense inflammatory priming by LB in this rat strain. Even though E.coli- and/or Enterococcus spp.-induced rise in H2O2 release in vitro was abrogated by LB in cells from both rat strains, LB prevented microbiota-induced increase in NO/urea ratio only in cells from AO and augmented it in cells from DA rats. Thus, the immunomodulatory properties may not be constant for particular probiotic bacteria, but shaped by innate immunity of the host.
Collapse
Affiliation(s)
- Stanislava Stanojević
- Immunology Research Centre "Branislav Janković, Institute of Virology, Vaccines and Sera "Torlak, Belgrade, Serbia. .,Immunology Research Centre "Branislav Janković, Institute of Virology, Vaccines and Sera "Torlak, Belgrade, Serbia.
| | - Veljko Blagojević
- Immunology Research Centre "Branislav Janković, Institute of Virology, Vaccines and Sera "Torlak, Belgrade, Serbia
| | - Ivana Ćuruvija
- Immunology Research Centre "Branislav Janković, Institute of Virology, Vaccines and Sera "Torlak, Belgrade, Serbia
| | - Vesna Vujić
- Department of Chemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
36
|
Induri SNR, Kansara P, Thomas SC, Xu F, Saxena D, Li X. The Gut Microbiome, Metformin, and Aging. Annu Rev Pharmacol Toxicol 2021; 62:85-108. [PMID: 34449247 DOI: 10.1146/annurev-pharmtox-051920-093829] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Metformin has been extensively used for the treatment of type 2 diabetes, and it may also promote healthy aging. Despite its widespread use and versatility, metformin's mechanisms of action remain elusive. The gut typically harbors thousands of bacterial species, and as the concentration of metformin is much higher in the gut as compared to plasma, it is plausible that microbiome-drug-host interactions may influence the functions of metformin. Detrimental perturbations in the aging gut microbiome lead to the activation of the innate immune response concomitant with chronic low-grade inflammation. With the effectiveness of metformin in diabetes and antiaging varying among individuals, there is reason to believe that the gut microbiome plays a role in the efficacy of metformin. Metformin has been implicated in the promotion and maintenance of a healthy gut microbiome and reduces many age-related degenerative pathologies. Mechanistic understanding of metformin in the promotion of a healthy gut microbiome and aging will require a systems-level approach. Expected final online publication date for the Annual Review of Pharmacology and Toxicology, Volume 62 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Sri Nitya Reddy Induri
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA;
| | - Payalben Kansara
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA;
| | - Scott C Thomas
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA;
| | - Fangxi Xu
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA;
| | - Deepak Saxena
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA; .,Department of Surgery, New York University School of Medicine, New York, NY 10016, USA
| | - Xin Li
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA;
| |
Collapse
|
37
|
Association of Polygenic Risk Score and Bacterial Toxins at Screening Colonoscopy with Colorectal Cancer Progression: A Multicenter Case-Control Study. Toxins (Basel) 2021; 13:toxins13080569. [PMID: 34437440 PMCID: PMC8402601 DOI: 10.3390/toxins13080569] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/06/2021] [Accepted: 08/11/2021] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) is a leading cause of cancer death worldwide, and its incidence is correlated with infections, chronic inflammation, diet, and genetic factors. An emerging aspect is that microbial dysbiosis and chronic infections triggered by certain bacteria can be risk factors for tumor progression. Recent data suggest that certain bacterial toxins implicated in DNA attack or in proliferation, replication, and death can be risk factors for insurgence and progression of CRC. In this study, we recruited more than 300 biopsy specimens from people undergoing colonoscopy, and we analyzed to determine whether a correlation exists between the presence of bacterial genes coding for toxins possibly involved in CRC onset and progression and the different stages of CRC. We also analyzed to determine whether CRC-predisposing genetic factors could contribute to bacterial toxins response. Our results showed that CIF toxin is associated with polyps or adenomas, whereas pks+ seems to be a predisposing factor for CRC. Toxins from Escherichia coli as a whole have a higher incidence rate in adenocarcinoma patients compared to controls, whereas Bacteroides fragilis toxin does not seem to be associated with pre-cancerous nor with cancerous lesions. These results have been obtained irrespectively of the presence of CRC-risk loci.
Collapse
|
38
|
Role of RONS and eIFs in Cancer Progression. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5522054. [PMID: 34285764 PMCID: PMC8275427 DOI: 10.1155/2021/5522054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 04/19/2021] [Accepted: 05/14/2021] [Indexed: 12/05/2022]
Abstract
Various research works have piled up conflicting evidence questioning the effect of oxidative stress in cancer. Reactive oxygen and nitrogen species (RONS) are the reactive radicals and nonradical derivatives of oxygen and nitrogen. RONS can act as a double-edged weapon. On the one hand, RONS can promote cancer initiation through activating certain signal transduction pathways that direct proliferation, survival, and stress resistance. On the other hand, they can mitigate cancer progression via their resultant oxidative stress that causes many cancer cells to die, as some recent studies have proposed that high RONS levels can limit the survival of cancer cells during certain phases of cancer development. Similarly, eukaryotic translation initiation factors are key players in the process of cellular transformation and tumorigenesis. Dysregulation of such translation initiation factors in the form of overexpression, downregulation, or phosphorylation is associated with cancer cell's altering capability of survival, metastasis, and angiogenesis. Nonetheless, eIFs can affect tumor age-related features. Data shows that alternating the eukaryotic translation initiation apparatus can impact many downstream cellular signaling pathways that directly affect cancer development. Hence, researchers have been conducting various experiments towards a new trajectory to find novel therapeutic molecular targets to improve the efficacy of anticancer drugs as well as reduce their side effects, with a special focus on oxidative stress and initiation of translation to harness their effect in cancer development. An increasing body of scientific evidence recently links oxidative stress and translation initiation factors to cancer-related signaling pathways. Therefore, in this review, we present and summarize the recent findings in this field linking certain signaling pathways related to tumorigeneses such as MAPK and PI3K, with either RONS or eIFs.
Collapse
|
39
|
Gatekeepers of the Gut: The Roles of Proteasomes at the Gastrointestinal Barrier. Biomolecules 2021; 11:biom11070989. [PMID: 34356615 PMCID: PMC8301830 DOI: 10.3390/biom11070989] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 12/24/2022] Open
Abstract
The gut epithelial barrier provides the first line of defense protecting the internal milieu from the environment. To circumvent the exposure to constant challenges such as pathogenic infections and commensal bacteria, epithelial and immune cells at the gut barrier require rapid and efficient means to dynamically sense and respond to stimuli. Numerous studies have highlighted the importance of proteolysis in maintaining homeostasis and adapting to the dynamic changes of the conditions in the gut environment. Primarily, proteolytic activities that are involved in immune regulation and inflammation have been examined in the context of the lysosome and inflammasome activation. Yet, the key to cellular and tissue proteostasis is the ubiquitin–proteasome system, which tightly regulates fundamental aspects of inflammatory signaling and protein quality control to provide rapid responses and protect from the accumulation of proteotoxic damage. In this review, we discuss proteasome-dependent regulation of the gut and highlight the pathophysiological consequences of the disarray of proteasomal control in the gut, in the context of aberrant inflammatory disorders and tumorigenesis.
Collapse
|
40
|
Wang H, Banerjee N, Liang Y, Wang G, Hoffman KL, Khan MF. Gut microbiome-host interactions in driving environmental pollutant trichloroethene-mediated autoimmunity. Toxicol Appl Pharmacol 2021; 424:115597. [PMID: 34051218 DOI: 10.1016/j.taap.2021.115597] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/18/2021] [Accepted: 05/24/2021] [Indexed: 12/15/2022]
Abstract
Trichloroethene (TCE), a widely used industrial solvent, is associated with the development of autoimmune diseases (ADs), including systemic lupus erythematosus and autoimmune hepatitis. Increasing evidence support a linkage between altered gut microbiome composition and the onset of ADs. However, it is not clear how gut microbiome contributes to TCE-mediated autoimmunity, and initial triggers for microbiome-host interactions leading to systemic autoimmune responses remain unknown. To achieve this, female MRL+/+ mice were treated with 0.5 mg/ml TCE for 52 weeks and fecal samples were subjected to 16S rRNA sequencing to determine the microbiome composition. TCE exposure resulted in distinct bacterial community revealed by β-diversity analysis. Notably, we observed reduction in Lactobacillaceae, Rikenellaceae and Bifidobacteriaceae families, and enrichment of Akkermansiaceae and Lachnospiraceae families after TCE exposure. We also observed significantly increased colonic oxidative stress and inflammatory markers (CD14 and IL-1β), and decreased tight junction proteins (ZO-2, occludin and claudin-3). These changes were associated with increases in serum antinuclear and anti-smooth muscle antibodies and cytokines (IL-6 and IL-12), together with increased PD1 + CD4+ T cells in TCE-exposed spleen and liver tissues. Importantly, fecal microbiota transplantation (FMT) using feces from TCE-treated mice to antibiotics-treated mice induced increased anti-dsDNA antibodies and hepatic CD4+ T cell infiltration in the recipient mice. Our studies thus delineate how imbalance in gut microbiome and mucosal redox status together with gut inflammatory response and permeability changes could be the key factors in contributing to TCE-mediated ADs. Furthermore, FMT studies provide a solid support to a causal role of microbiome in TCE-mediated autoimmunity.
Collapse
Affiliation(s)
- Hui Wang
- Department of Pathology, University of Texas Medical Branch, TX, United States of America
| | - Nivedita Banerjee
- Department of Pathology, University of Texas Medical Branch, TX, United States of America
| | - Yuejin Liang
- Department of Microbiology and Immunology, University of Texas Medical Branch, TX, United States of America
| | - Gangduo Wang
- Department of Pathology, University of Texas Medical Branch, TX, United States of America
| | - Kristi L Hoffman
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - M Firoze Khan
- Department of Pathology, University of Texas Medical Branch, TX, United States of America.
| |
Collapse
|
41
|
Tavakoli P, Vollmer-Conna U, Hadzi-Pavlovic D, Grimm MC. A Review of Inflammatory Bowel Disease: A Model of Microbial, Immune and Neuropsychological Integration. Public Health Rev 2021; 42:1603990. [PMID: 34692176 PMCID: PMC8386758 DOI: 10.3389/phrs.2021.1603990] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 04/01/2021] [Indexed: 12/11/2022] Open
Abstract
Objective: Inflammatory bowel diseases (IBDs) are complex chronic inflammatory disorders of the gastro-intestinal (GI) tract with uncertain etiology. IBDs comprise two idiopathic disorders: Crohn's disease (CD) and ulcerative colitis (UC). The aetiology, severity and progression of such disorders are still poorly understood but thought to be influenced by multiple factors (including genetic, environmental, immunological, physiological, psychological factors and gut microbiome) and their interactions. The overarching aim of this review is to evaluate the extent and nature of the interrelationship between these factors with the disease course. A broader conceptual and longitudinal framework of possible neuro-visceral integration, core microbiome analysis and immune modulation assessment may be useful in accurately documenting and characterizing the nature and temporal continuity of crosstalk between these factors and the role of their interaction (s) in IBD disease activity. Characterization of these interactions holds the promise of identifying novel diagnostic, interventions, and therapeutic strategies. Material and Methods: A search of published literature was conducted by exploring PubMed, EMBASE, MEDLINE, Medline Plus, CDSR library databases. Following search terms relating to key question were set for the search included: "Inflammatory bowel diseases," "gut microbiota," "psychological distress and IBD," "autonomic reactivity and IBD," "immune modulation," "chronic inflammation," "gut inflammation," "enteric nervous system," "gut nervous system," "Crohn's disease," "Ulcerative colitis", "depression and IBD", "anxiety and IBD", "quality of life in IBD patients," "relapse in IBDs," "remission in IBDs," "IBD disease activity," "brain-gut-axis," "microbial signature in IBD," "validated questionnaires in IBD," "IBD activity indices," "IBD aetiology," "IBDs and stress," "epidemiology of IBDs", "autonomic nervous system and gut inflammation", "IBD and environment," "genetics of IBDs," "pathways of immune response in IBDs," "sleep disturbances in IBD," "hypothalamic-pituitary-adrenal axis (HPA)," "sympatho-adrenal axis," "CNS and its control of gut function" "mucosal immune response," "commensal and pathogenic bacteria in the gut," "innate and adaptive immunity." Studies evaluating any possible associations between gut microbiome, psychological state, immune modulation, and autonomic function with IBDs were identified. Commonly cited published literatures with high quality research methodology/results and additional articles from bibliographies of recovered papers were examined and included where relevant. Results: Although there is a substantial literature identifying major contributing factors with IBD, there has been little attempt to integrate some factors over time and assess their interplay and relationship with IBD disease activity. Such contributing factors include genetic and environmental factors, gut microbiota composition and function, physiological factors, psychological state and gut immune response. Interdependences are evident across psychological and biological factors and IBD disease activity. Although from the available evidence, it is implausible that a single explanatory model could elucidate the interplay between such factors and the disease course as well as the sequence of the effect during the pathophysiology of IBD. Conclusion: Longitudinal monitoring of IBD patients and integrating data related to the contributing/risk factors including psychological state, physiological conditions, inflammatory/immune modulations, and microbiome composition/function, could help to explain how major factors associate and interrelate leading to exacerbation of symptoms and disease activity. Identifying the temporal trajectory of biological and psychosocial disturbances may also help to assess their effects and interdependence on individuals' disease status. Moreover, this allows greater insight into understanding the temporal progressions of subclinical events as potential ground for disease severity in IBD. Furthermore, understanding the interaction between these risk factors may help better interventions in controlling the disease, reducing the costs related to disease management, further implications for clinical practice and research approaches in addition to improving patients' mental health and quality of life.
Collapse
Affiliation(s)
- P. Tavakoli
- St George and Sutherland Clinical School, Sydney, NSW, Australia
| | - U. Vollmer-Conna
- School of Psychiatry, University of New South Wales, Sydney, Australia
| | - D. Hadzi-Pavlovic
- School of Psychiatry, University of New South Wales, Sydney, Australia
| | - M. C. Grimm
- St George and Sutherland Clinical School, Sydney, NSW, Australia
| |
Collapse
|
42
|
Rocks T, West M, Hockey M, Aslam H, Lane M, Loughman A, Jacka FN, Ruusunen A. Possible use of fermented foods in rehabilitation of anorexia nervosa: the gut microbiota as a modulator. Prog Neuropsychopharmacol Biol Psychiatry 2021; 107:110201. [PMID: 33307114 DOI: 10.1016/j.pnpbp.2020.110201] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 11/23/2020] [Accepted: 12/02/2020] [Indexed: 12/15/2022]
Abstract
Anorexia nervosa is a serious psychiatric disorder with high morbidity and mortality rate. Evidence for the optimal psychopharmacological approach to managing the disorder remains limited, with nutritional treatment, focused on weight restoration through the consumption of high energy diet, regarded as one of the fundamental steps in treatment. The human gut microbiome is increasingly recognised for its proposed role in gastrointestinal, metabolic, immune and mental health, all of which may be compromised in individuals with anorexia nervosa. Dietary intake plays an important role in shaping gut microbiota composition, whilst the use of fermented foods, foods with potential psychobiotic properties that deliver live bacteria, bacterial metabolites, prebiotics and energy, have been discussed to a lesser extent. However, fermented foods are of increasing interest due to their potential capacity to affect gut microbiota composition, provide beneficial bacterial metabolites, and confer beneficial outcomes to host health. This review provides an overview of the role of the gut microbiota in relation to the disease pathology in anorexia nervosa and especially focuses on the therapeutic potential of fermented foods, proposed here as a recommended addition to the current nutritional treatment protocols warranting further investigation.
Collapse
Affiliation(s)
- Tetyana Rocks
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Geelong, Australia.
| | - Madeline West
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Geelong, Australia
| | - Meghan Hockey
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Geelong, Australia
| | - Hajara Aslam
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Geelong, Australia
| | - Melissa Lane
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Geelong, Australia
| | - Amy Loughman
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Geelong, Australia
| | - Felice N Jacka
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Geelong, Australia; Centre for Adolescent Health, Murdoch Children's Research Institute, VIC, Australia; Black Dog Institute, NSW, Australia; James Cook University, QLD; Australia
| | - Anu Ruusunen
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Geelong, Australia; Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland; Department of Psychiatry, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
43
|
Li C, Zhang L, Qian D, Cheng M, Hu H, Hong Z, Cui Y, Yu H, Wang Q, Zhu J, Meng W, Xu JF, Sun Y, Zhang P, Wang C. RNF111-facilitated neddylation potentiates cGAS-mediated antiviral innate immune response. PLoS Pathog 2021; 17:e1009401. [PMID: 33720974 PMCID: PMC7959372 DOI: 10.1371/journal.ppat.1009401] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 02/17/2021] [Indexed: 12/15/2022] Open
Abstract
The cytosolic DNA sensor cyclic GMP-AMP (cGAMP) synthetase (cGAS) has emerged as a fundamental component fueling the anti-pathogen immunity. Because of its pivotal role in initiating innate immune response, the activity of cGAS must be tightly fine-tuned to maintain immune homeostasis in antiviral response. Here, we reported that neddylation modification was indispensable for appropriate cGAS-STING signaling activation. Blocking neddylation pathway using neddylation inhibitor MLN4924 substantially impaired the induction of type I interferon and proinflammatory cytokines, which was selectively dependent on Nedd8 E2 enzyme Ube2m. We further found that deficiency of the Nedd8 E3 ligase Rnf111 greatly attenuated DNA-triggered cGAS activation while not affecting cGAMP induced activation of STING, demonstrating that Rnf111 was the Nedd8 E3 ligase of cGAS. By performing mass spectrometry, we identified Lys231 and Lys421 as essential neddylation sites in human cGAS. Mechanistically, Rnf111 interacted with and polyneddylated cGAS, which in turn promoted its dimerization and enhanced the DNA-binding ability, leading to proper cGAS-STING pathway activation. In the same line, the Ube2m or Rnf111 deficiency mice exhibited severe defects in innate immune response and were susceptible to HSV-1 infection. Collectively, our study uncovered a vital role of the Ube2m-Rnf111 neddylation axis in promoting the activity of the cGAS-STING pathway and highlighted the importance of neddylation modification in antiviral defense.
Collapse
Affiliation(s)
- Chenhui Li
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Lele Zhang
- Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Dong Qian
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Mingxing Cheng
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Haiyang Hu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Ze Hong
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Ye Cui
- Division of Immunology, The Boston Children’s Hospital, Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Huansha Yu
- Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Quanyi Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Juanjuan Zhu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Wei Meng
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Jin-fu Xu
- Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yi Sun
- Cancer Institute of the 2 affiliated hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- * E-mail: (YS); (PZ); (CW)
| | - Peng Zhang
- Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- * E-mail: (YS); (PZ); (CW)
| | - Chen Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
- * E-mail: (YS); (PZ); (CW)
| |
Collapse
|
44
|
Wierzba W, Karnafel W, Śliwczyński A, Pinkas J, Gujski M. Diabetes mellitus and congestive heart failure: the prevalence of congestive heart failure in patients with and without diabetes in Poland. Arch Med Sci 2021; 17:646-651. [PMID: 34025834 PMCID: PMC8130468 DOI: 10.5114/aoms.2018.74261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 02/26/2018] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Diabetes mellitus is a systemic disease and has a negative effect on the cardiovascular system. This paper aimed to present a retrospective analysis of morbidity associated with heart failure in subgroups of patients with and without diabetes in Poland in 2012. MATERIAL AND METHODS Data from the National Health Fund were used for the study. In general, 656,937 patients with heart failure, including 281,538 males and 375,354 females, were studied. In this population, additionally, 201,043 patients with heart failure (main diagnosis) and diabetes were studied, including 82,117 males and 118,926 females. RESULTS The mean index of morbidity associated with heart failure in the whole subpopulation of diabetes patients was 9.03%; 8.42% for males and 9.50% for females. Morbidity associated with heart failure in the population of patients diagnosed with diabetes in Poland in 2012 was seven times higher compared to morbidity associated with heart failure in non-diabetes patients. Morbidity associated with heart failure in females was significantly higher compared to morbidity in males in the whole population, in both the subpopulations of patients with and without diabetes. CONCLUSIONS Diabetes mellitus significantly increases risk of heart failure in both women and men. The risk is significantly high after the age of 60 years and higher in females.
Collapse
Affiliation(s)
- Waldemar Wierzba
- Department of Public Health, University of Humanities and Economics, Lodz, Poland
| | - Waldemar Karnafel
- Department of Public Health, University of Humanities and Economics, Lodz, Poland
| | - Andrzej Śliwczyński
- Department of Public Health, University of Humanities and Economics, Lodz, Poland
- National Health Fund, Central Office, Warsaw, Poland
| | - Jarosław Pinkas
- School of Public Health, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Mariusz Gujski
- Department of Prevention of Environmental Hazards and Allergology, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
45
|
Kim E, Cho S. CNS and peripheral immunity in cerebral ischemia: partition and interaction. Exp Neurol 2021; 335:113508. [PMID: 33065078 PMCID: PMC7750306 DOI: 10.1016/j.expneurol.2020.113508] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/28/2020] [Accepted: 10/08/2020] [Indexed: 02/07/2023]
Abstract
Stroke elicits excessive immune activation in the injured brain tissue. This well-recognized neural inflammation in the brain is not just an intrinsic organ response but also a result of additional intricate interactions between infiltrating peripheral immune cells and the resident immune cells in the affected areas. Given that there is a finite number of immune cells in the organism at the time of stroke, the partitioned immune systems of the central nervous system (CNS) and periphery must appropriately distribute the limited pool of immune cells between the two domains, mounting a necessary post-stroke inflammatory response by supplying a sufficient number of immune cells into the brain while maintaining peripheral immunity. Stroke pathophysiology has mainly been neurocentric in focus, but understanding the distinct roles of the CNS and peripheral immunity in their concerted action against ischemic insults is crucial. This review will discuss stroke-induced influences of the peripheral immune system on CNS injury/repair and of neural inflammation on peripheral immunity, and how comorbidity influences each.
Collapse
Affiliation(s)
- Eunhee Kim
- Vivian L. Smith Department of Neurosurgery at University of Texas Health Science Center at Houston, Houston, TX, United States of America
| | - Sunghee Cho
- Burke Neurological Institute, White Plains, NY, United States of America; Feil Brain Mind Research Institute, Weill Cornell Medicine, New York, NY, United States of America.
| |
Collapse
|
46
|
The Protective Role of Probiotics against Colorectal Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8884583. [PMID: 33488940 PMCID: PMC7803265 DOI: 10.1155/2020/8884583] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/02/2020] [Accepted: 11/26/2020] [Indexed: 02/06/2023]
Abstract
Colorectal cancer (CRC) is the fourth leading cause of cancer-related deaths worldwide and a major global public health problem. With the rapid development of the economy, the incidence of CRC has increased linearly. Accumulating evidence indicates that changes in the gut microenvironment, such as undesirable changes in the microbiota composition, provide favorable conditions for intestinal inflammation and shaping the tumor growth environment, whereas administration of certain probiotics can reverse this situation to a certain extent. This review summarizes the roles of probiotics in the regulation of CRC, such as enhancing the immune barrier, regulating the intestinal immune state, inhibiting pathogenic enzyme activity, regulating CRC cell proliferation and apoptosis, regulating redox homeostasis, and reprograming intestinal microbial composition. Abundant studies have provided a theoretical foundation for the roles of probiotics in CRC prevention and treatment, but their mechanisms of action remain to be investigated, and further clinical trials are warranted for the application of probiotics in the target population.
Collapse
|
47
|
Wang Y, Chen Y, Zhang X, Lu Y, Chen H. New insights in intestinal oxidative stress damage and the health intervention effects of nutrients: A review. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.104248] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
48
|
An evaluation of the effects of probiotics on tumoral necrosis factor (TNF-α) signaling and gene expression. Cytokine Growth Factor Rev 2020; 57:27-38. [PMID: 33162326 DOI: 10.1016/j.cytogfr.2020.10.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 10/06/2020] [Accepted: 10/09/2020] [Indexed: 12/16/2022]
Abstract
The search for functional foods containing probiotics has been growing due to numerous benefits they provide to health, such as modulation of the immune system and of the anti-inflammatory activity by inhibiting the release of pro-inflammatory cytokines, such as TNF-α. However, the mechanisms of actions of the probiotics responsible for this inhibition have not been completely explained so far. A better understanding of the interaction between probiotics and cell signaling pathways related to inflammatory processes shall help to prevent inflammatory bowel diseases. Therefore, the aim of this revision is to help understand the mechanisms of action of probiotics in cell signaling pathways that regulate TNF-α expression. Probiotics might act at different points of the MAPK pathway, on NF-kB, on proteasome activity, on Toll-like receptors, and on their regulators and stimuli. The present revision reaches the conclusion that probiotics act through multiple mechanisms, especially by inhibiting IkB phosphorylation and degradation, thus preventing the translocation of NF-kB. Effects are also shown to be strain-specific, and probiotics of the genus Lactobacillus are proved to play and essential role in anti-inflammatory activity.
Collapse
|
49
|
Is inflammageing influenced by the microbiota in the aged gut? A systematic review. Exp Gerontol 2020; 141:111079. [DOI: 10.1016/j.exger.2020.111079] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 08/02/2020] [Accepted: 08/28/2020] [Indexed: 12/21/2022]
|
50
|
Zou T, Zhang J. Diverse and pivotal roles of neddylation in metabolism and immunity. FEBS J 2020; 288:3884-3912. [PMID: 33025631 DOI: 10.1111/febs.15584] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/15/2020] [Accepted: 10/01/2020] [Indexed: 12/11/2022]
Abstract
Neddylation is one type of protein post-translational modification by conjugating a ubiquitin-like protein neural precursor cell-expressed developmentally downregulated protein 8 to substrate proteins via a cascade involving E1, E2, and E3 enzymes. The best-characterized substrates of neddylation are cullins, essential components of cullin-RING E3 ubiquitin-ligase complexes. The discovery of noncullin neddylation targets indicates that neddylation may have diverse biological functions. Indeed, neddylation has been implicated in various cellular processes including cell cycle progression, metabolism, immunity, and tumorigenesis. Here, we summarized the reported neddylation substrates and also discuss the functions of neddylation in the immune system and metabolism.
Collapse
Affiliation(s)
- Tao Zou
- Beijing Institute of Brain Sciences, China
| | | |
Collapse
|