151
|
Wang D, Zhu L, Liu H, Feng X, Zhang C, Liu B, Li T, Liu L, Chang H, Sun J, Yang L, Yang W. Altered gut metabolites and metabolic reprogramming involved in the pathogenesis of colitis-associated colorectal cancer and the transition of colon "inflammation to cancer". J Pharm Biomed Anal 2024; 253:116553. [PMID: 39486392 DOI: 10.1016/j.jpba.2024.116553] [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: 07/03/2024] [Revised: 10/27/2024] [Accepted: 10/27/2024] [Indexed: 11/04/2024]
Abstract
Colitis-associated colorectal cancer (CAC) is fatal and can develop spontaneously or as a complication of inflammatory bowel diseases. Although co-administration of azoxymethane/dextran sulfate sodium (AOM/DSS) is a classic method for CAC modeling, its limitations need to be addressed. Accordingly, we aimed to optimize the AOM/DSS model to study CAC extensively and further investigate its pathogenic mechanisms relative to microbiota and metabolism. We optimized the CAC model via a single or enhanced injection of AOM combined with different administration modes and varying DSS concentrations. Subsequently, the fecal-microbiota composition was examined using 16S RNA sequencing, and fecal-colon-metabolome profiles were evaluated via ultra-high performance liquid chromatography-mass spectrometry. Two interval injections of AOM combined with 1.5 % DSS-free drinking resulted in a high tumor formation rate, uniform tumor formation, and low mortality. Based on this model, we innovatively divided the pathogenesis of CAC into three stages, namely inflammation induction, proliferation initiation, and tumorigenesis, and examined the pathological characteristics in each stage. Gut microbial dysbiosis and metabolic alteration drove colorectal tumorigenesis by aggravating inflammation while promoting cell proliferation and carcinogenesis in mice. For the first time, we dynamically demonstrated the process of colon "inflammation to cancer" transformation and provided novel insights to clarify the role of amino acid metabolism in the formation of CAC.
Collapse
Affiliation(s)
- Dunfang Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Lin Zhu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Haifan Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xue Feng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Caijuan Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Bin Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Tao Li
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Li Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Hao Chang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Jingwei Sun
- Beijing University of Chinese Medicine, Beijing 100700, China
| | - Lei Yang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Weipeng Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| |
Collapse
|
152
|
Aierken A, Aizezi Y, Atabieke F, Rehaman M, Aierken M, Li SX. No bidirectional relationship between constipation and colorectal cancer in European and Asian populations: A Mendelian randomization study. Medicine (Baltimore) 2024; 103:e40206. [PMID: 39470562 PMCID: PMC11520989 DOI: 10.1097/md.0000000000040206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/23/2024] [Accepted: 10/04/2024] [Indexed: 10/30/2024] Open
Abstract
Traditional observational studies have reported a positive association between constipation and the risk of colorectal cancer (CRC). However, evidence from other approaches to pursue the causal relationship between constipation and CRC is scarce. In the study, 2-sample Mendelian randomization analysis was conducted to investigate the potential causal relationship between constipation and CRC. Analysis of the results showed that there was no causal association between constipation and CRC, either in European populations (CRC: odds ratio [OR] = 1.00, 95% confidence interval [CI] = 0.99-1.00, P = .49; rectal cancer: OR = 0.99, 95% CI = 0.99-1.00, P = .79) or in Asian populations (CRC: OR = 1.00, 95% CI = 0.99-1.01, P = .30). Also there was no inverse causal association between CRC and constipation, either in European populations (CRC: OR = 0.10, 95% CI = 2.76E-03-3.45, P = .20; rectal cancer: OR = 0.05, 95% CI = 9.14E-07-2.64E + 03, P = .59) or in Asian population (CRC: OR = 1.18, 95% CI = 0.92-1.52, P = .20), there was no horizontal diversity in the instrumental variables in the Mendelian randomization analyses of the present study (all F statistics >10), and no heterogeneity was found in the regression analyses. The findings from bidirectional 2-sample Mendelian randomization analyses indicate that there is no evidence of a bidirectional causal association between constipation and CRC. However, further investigation is warranted through additional clinical studies and trials to thoroughly explore the association between these 2 factors.
Collapse
Affiliation(s)
- Ailikamu Aierken
- Graduate School of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, China
- Department of General Surgery, Children’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Yierzhati Aizezi
- Department of Critical Care Medicine, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Falide Atabieke
- The second Department of Gastroenterology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Mayinuer Rehaman
- Department of Physiology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Munire Aierken
- Department of Disinfection and Vector-Borne Pathogen Control, Urumqi City Center for Disease Prevention and Control, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Shui-Xue Li
- Department of General Surgery, Children’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| |
Collapse
|
153
|
Hoffbeck C, Middleton DRML, Keall SN, Huang CM, Pas A, Irving K, Nelson NJ, Taylor MW. Limited gut bacterial response of tuatara (Sphenodon punctatus) to dietary manipulation and captivity. FEMS Microbiol Ecol 2024; 100:fiae141. [PMID: 39400705 PMCID: PMC11523620 DOI: 10.1093/femsec/fiae141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 09/26/2024] [Accepted: 10/12/2024] [Indexed: 10/15/2024] Open
Abstract
The bacteria of a host's digestive tract play crucial roles in digestion and pathogen resistance. Hosts living in captivity often have more human interaction and antibiotic use, in addition to differences in diet and environment, compared to their wild counterparts. Consequently, wild and captive animals frequently harbour different bacterial communities. We tested whether diversity of diet provided in captivity shifts the gut bacteria of tuatara, an endemic New Zealand reptile, at three captive sites, and examined how the gut community of these tuatara compares to those in the wild. Dietary manipulation did not cause a strong overall shift in tuatara gut bacteria, but individual tuatara did experience bacterial shifts during manipulation, which subsequently reverted after manipulation. We found that Bacteroides, a genus common in most vertebrate guts but rare in tuatara, increased significantly in the gut during manipulation, then decreased post-manipulation. Finally, the gut bacteria of captive tuatara significantly differed from those of wild tuatara, though most of the dominant bacterial genera found in wild tuatara persisted in captive tuatara. This work represents a first investigation of the captive tuatara bacterial community and establishes the sensitivity of the gut community to dietary manipulation and captivity for this relict reptile.
Collapse
Affiliation(s)
- Carmen Hoffbeck
- School of Biological Sciences, University of Auckland, Auckland 1010, New Zealand
| | | | - Susan N Keall
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
| | | | - An Pas
- Auckland Zoo, Auckland 1022, New Zealand
| | - Kate Irving
- Wellington Zoo, Wellington 6021, New Zealand
| | - Nicola J Nelson
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Michael W Taylor
- School of Biological Sciences, University of Auckland, Auckland 1010, New Zealand
| |
Collapse
|
154
|
Song HA, Jang SY, Park MJ, Kim SW, Kang CG, Lee JH, Kim HJ, Kim J, Lee JK, Chung KS, Lee KT. Immunostimulation Signaling via Toll-like Receptor 2 Activation: A Molecular Mechanism of Lactococcus lactis OTG1204 In Vitro and In Vivo. Nutrients 2024; 16:3629. [PMID: 39519462 PMCID: PMC11547582 DOI: 10.3390/nu16213629] [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: 09/26/2024] [Revised: 10/18/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
INTRODUCTION The immune system's defense against pathogens involves innate and adaptive responses, crucial in maintaining overall health. Immunosuppressed states render individuals more susceptible to potential diseases, indicating the need for effective strategies to bolster immune functions. OBJECTIVES Although the immunostimulatory effects of various probiotics have been studied, the specific effects and molecular mechanisms of Lactococcus lactis OTG1204 (OTG1204) remain unknown. In this study, the aim was to investigate the molecular mechanisms of OTG1204 in RAW 264.7 macrophages, the key effector cells of the innate immune system involved in host defense and inflammatory responses. Additionally, in this study, the effects of OTG1204 on cyclophosphamide (CTX)-induced immunosuppression states were investigated, thereby demonstrating its potential as an immune stimulant. METHODS To assess the macrophage activation ability and underlying mechanisms of OTG1204, RAW 264.7 cells were utilized with transfection, enzyme-linked immunosorbent assay, and quantitative real-time PCR analyses. Furthermore, to evaluate the immunostimulatory effects under immunosuppressed conditions, CTX-induced immunosuppression mice model was employed, and analyses were performed using hematoxylin and eosin staining, flow cytometry, and microbiota examination. RESULTS OTG1204 activated RAW 264.7 macrophages, leading to increased production of nitric oxide, prostaglandin E2, and cytokines. This immune activation was mediated through the upregulation of toll-like receptor 2, which subsequently activated the nuclear factor-κB (NF-kB) and mitogen-activated protein kinase (MAPK)/activator protein 1 (AP-1) pathways, thereby stimulating the immune response. In CTX-treated mice, OTG1204 recovered body weight, spleen, and mesenteric lymph node indices, and natural killer cell activity. It re-established populations of innate and adaptive immune cells and activated T cells to secrete cytokines. We also examined the gut barrier integrity and microbiota composition to assess OTG1204's impact on intestinal health, as these factors play a significant role in immune enhancement. OTG1204 enhanced gut barrier integrity by upregulating mucin 2 and tight junction proteins and modulated the gut microbiota by restoring the Firmicutes/Bacteroidetes balance and reducing the abundance of Actinobacteria and Tenericutes. CONCLUSION These results suggest that OTG1204 may serve as an effective probiotic for immune enhancement and gut health management by targeting the NF-κB and MAPK/AP-1 pathways, with minimal side effects.
Collapse
Affiliation(s)
- Hyeon-A Song
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; (H.-A.S.); (S.-Y.J.); (M.-J.P.); (K.-S.C.)
- Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Seo-Yun Jang
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; (H.-A.S.); (S.-Y.J.); (M.-J.P.); (K.-S.C.)
- Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Min-Ji Park
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; (H.-A.S.); (S.-Y.J.); (M.-J.P.); (K.-S.C.)
- Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Seung Wook Kim
- Ottogi Research Center, Anyang 14060, Republic of Korea; (S.W.K.); (C.G.K.); (J.H.L.); (H.-J.K.); (J.K.)
| | - Choon Gil Kang
- Ottogi Research Center, Anyang 14060, Republic of Korea; (S.W.K.); (C.G.K.); (J.H.L.); (H.-J.K.); (J.K.)
| | - Joo Hyun Lee
- Ottogi Research Center, Anyang 14060, Republic of Korea; (S.W.K.); (C.G.K.); (J.H.L.); (H.-J.K.); (J.K.)
| | - Hye-Jin Kim
- Ottogi Research Center, Anyang 14060, Republic of Korea; (S.W.K.); (C.G.K.); (J.H.L.); (H.-J.K.); (J.K.)
| | - Jiheon Kim
- Ottogi Research Center, Anyang 14060, Republic of Korea; (S.W.K.); (C.G.K.); (J.H.L.); (H.-J.K.); (J.K.)
| | - Jong Kil Lee
- Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Kyung-Sook Chung
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; (H.-A.S.); (S.-Y.J.); (M.-J.P.); (K.-S.C.)
| | - Kyung-Tae Lee
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; (H.-A.S.); (S.-Y.J.); (M.-J.P.); (K.-S.C.)
- Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea;
| |
Collapse
|
155
|
Basu A, Adams AND, Degnan PH, Vanderpool CK. Determinants of raffinose family oligosaccharide use in Bacteroides species. J Bacteriol 2024; 206:e0023524. [PMID: 39330254 PMCID: PMC11501099 DOI: 10.1128/jb.00235-24] [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: 06/03/2024] [Accepted: 09/06/2024] [Indexed: 09/28/2024] Open
Abstract
Bacteroides species are successful colonizers of the human colon and can utilize a wide variety of complex polysaccharides and oligosaccharides that are indigestible by the host. To do this, they use enzymes encoded in polysaccharide utilization loci (PULs). While recent work has uncovered the PULs required for the use of some polysaccharides, how Bacteroides utilize smaller oligosaccharides is less well studied. Raffinose family oligosaccharides (RFOs) are abundant in plants, especially legumes, and consist of variable units of galactose linked by α-1,6 bonds to a sucrose (glucose α-1-β-2 fructose) moiety. Previous work showed that an α-galactosidase, BT1871, is required for RFO utilization in Bacteroides thetaiotaomicron. Here, we identify two different types of mutations that increase BT1871 mRNA levels and improve B. thetaiotaomicron growth on RFOs. First, a novel spontaneous duplication of BT1872 and BT1871 places these genes under the control of a ribosomal promoter, driving high BT1871 transcription. Second, nonsense mutations in a gene encoding the PUL24 anti-sigma factor likewise increase BT1871 transcription. We then show that hydrolases from PUL22 work together with BT1871 to break down the sucrose moiety of RFOs and determine that the master regulator of carbohydrate utilization (BT4338) plays a role in RFO utilization in B. thetaiotaomicron. Examining the genomes of other Bacteroides species, we found homologs of BT1871 in a subset and showed that representative strains of species with a BT1871 homolog grew better on melibiose than species that lack a BT1871 homolog. Altogether, our findings shed light on how an important gut commensal utilizes an abundant dietary oligosaccharide. IMPORTANCE The gut microbiome is important in health and disease. The diverse and densely populated environment of the gut makes competition for resources fierce. Hence, it is important to study the strategies employed by microbes for resource usage. Raffinose family oligosaccharides are abundant in plants and are a major source of nutrition for the microbiota in the colon since they remain undigested by the host. Here, we study how the model commensal organism, Bacteroides thetaiotaomicron utilizes raffinose family oligosaccharides. This work highlights how an important member of the microbiota uses an abundant dietary resource.
Collapse
Affiliation(s)
- Anubhav Basu
- Department of Microbiology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Amanda N. D. Adams
- Department of Microbiology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Patrick H. Degnan
- Department of Microbiology and Plant Pathology, University of California Riverside, Riverside, California, USA
| | - Carin K. Vanderpool
- Department of Microbiology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
156
|
Sánchez MC, Hernández P, Velapatiño Á, Cuba E, Ciudad MJ, Collado L. Illumina Sequencing in Conjunction with Propidium Monoazide to Identify Live Bacteria After Antiseptic Treatment in a Complex Oral Biofilm: A Study Using an Ex Vivo Supragingival Biofilm Model. Antibiotics (Basel) 2024; 13:1000. [PMID: 39596695 PMCID: PMC11591144 DOI: 10.3390/antibiotics13111000] [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: 09/27/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: The evaluation of the efficacy of antibacterial treatments in complex oral ecosystems is limited by the inability to differentiate live from dead bacteria using omic techniques. The objective of this study was therefore to assess the ability of the combination of the 16S rRNA Illumina sequencing methodology and the action of propidium monoazide (PMA) to study viable bacterial profiles in oral biofilms after exposure to an antiseptic compound. Methods: Cariogenic supragingival biofilms were developed in an ex vivo model for 96 h, using saliva from healthy volunteers. The biofilms were treated with 0.12% chlorhexidine (CHX) combined with 0.05% cetylpyridinium chloride (CPC), for 60 s, using phosphate buffered saline as a control. After exposure, each biofilm was treated or not with PMA to then extract the bacterial DNA, quantify it by Qubit, quantify the bacterial population using qPCR, and perform the metataxonomic study of the samples using Illumina 16S rRNA sequencing. Results: A significantly lower DNA concentration in the PMA-treated biofilms (p < 0.05 compared with those not exposed to PMA) was observed. The viable bacterial count obtained by qPCR differed significantly from the total bacterial count in the biofilm samples exposed to the antiseptic (p < 0.05). The viable microbiome differed significantly from the total bacterial profile of the samples treated with CHX/CPC after exposure to PMA (p < 0.05 at the α- and β-diversity levels). Conclusions: The combination of Illumina 16S rRNA sequencing and PMA helps solve the inability to evaluate the efficacy of antibacterial treatments in the bacterial profile of complex ecosystems such as oral biofilms.
Collapse
Affiliation(s)
- María C. Sánchez
- Department of Medicine, Faculty of Medicine, University Complutense, 28040 Madrid, Spain; (M.C.S.); (P.H.); (Á.V.); (E.C.); (L.C.)
- GINTRAMIS Research Group (Translational Research Group on Microbiota and Health), Faculty of Medicine, University Complutense, 28040 Madrid, Spain
| | - Paola Hernández
- Department of Medicine, Faculty of Medicine, University Complutense, 28040 Madrid, Spain; (M.C.S.); (P.H.); (Á.V.); (E.C.); (L.C.)
| | - Ángela Velapatiño
- Department of Medicine, Faculty of Medicine, University Complutense, 28040 Madrid, Spain; (M.C.S.); (P.H.); (Á.V.); (E.C.); (L.C.)
| | - Eber Cuba
- Department of Medicine, Faculty of Medicine, University Complutense, 28040 Madrid, Spain; (M.C.S.); (P.H.); (Á.V.); (E.C.); (L.C.)
| | - María J. Ciudad
- Department of Medicine, Faculty of Medicine, University Complutense, 28040 Madrid, Spain; (M.C.S.); (P.H.); (Á.V.); (E.C.); (L.C.)
- GINTRAMIS Research Group (Translational Research Group on Microbiota and Health), Faculty of Medicine, University Complutense, 28040 Madrid, Spain
| | - Luis Collado
- Department of Medicine, Faculty of Medicine, University Complutense, 28040 Madrid, Spain; (M.C.S.); (P.H.); (Á.V.); (E.C.); (L.C.)
- GINTRAMIS Research Group (Translational Research Group on Microbiota and Health), Faculty of Medicine, University Complutense, 28040 Madrid, Spain
| |
Collapse
|
157
|
Grönroos M, Jumpponen A, Roslund MI, Nurminen N, Oikarinen S, Parajuli A, Laitinen OH, Cinek O, Kramna L, Rajaniemi J, Hyöty H, Puhakka R, Sinkkonen A. Using patterns of shared taxa to infer bacterial dispersal in human living environment in urban and rural areas. Appl Environ Microbiol 2024; 90:e0090324. [PMID: 39230286 PMCID: PMC11498140 DOI: 10.1128/aem.00903-24] [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: 05/31/2024] [Accepted: 07/26/2024] [Indexed: 09/05/2024] Open
Abstract
Contact with environmental microbial communities primes the human immune system. Factors determining the distribution of microorganisms, such as dispersal, are thus important for human health. Here, we used the relative number of bacteria shared between environmental and human samples as a measure of bacterial dispersal and studied these associations with living environment and lifestyles. We analyzed amplicon sequence variants (ASVs) of the V4 region of 16S rDNA gene from 347 samples of doormat dust as well as samples of saliva, skin swabs, and feces from 53 elderly people in urban and rural areas in Finland at three timepoints. We first enumerated the ASVs shared between doormat and one of the human sample types (i.e., saliva, skin swab, or feces) of each individual subject and calculated the shared ASVs as a proportion of all ASVs in the given sample type of that individual. We observed that the patterns for the proportions of shared ASVs differed among seasons and human sample type. In skin samples, there was a negative association between the proportion of shared ASVs and the coverage of built environment (a proxy for degree of urbanization), whereas in saliva data, this association was positive. We discuss these findings in the context of differing species pools in urban and rural environments. IMPORTANCE Understanding how environmental microorganisms reach and interact with humans is a key question when aiming to increase human contacts with natural microbiota. Few methods are suitable for studying microbial dispersal at relatively large spatial scales. Thus, we tested an indirect method and studied patterns of bacterial taxa that are shared between humans and their living environment.
Collapse
Affiliation(s)
- M. Grönroos
- Faculty of Biological and Environmental Sciences, University of Helsinki, Lahti, Finland
| | - A. Jumpponen
- Division of Biology and Ecological Genomics Institute, Kansas State University, Manhattan, Kansas, USA
| | - M. I. Roslund
- Natural Resources Institute Finland, Helsinki, Finland
| | - N. Nurminen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - S. Oikarinen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - A. Parajuli
- Faculty of Biological and Environmental Sciences, University of Helsinki, Lahti, Finland
- Department of Medicine, Karolinska Institutet, Huddinge, Sweden
| | - O. H. Laitinen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - O. Cinek
- Department of Medical Microbiology, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - L. Kramna
- Department of Medical Microbiology, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - J. Rajaniemi
- Faculty of Built Environment, Tampere University, Tampere, Finland
| | - H. Hyöty
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Fimlab Laboratories, Pirkanmaa Hospital District, Tampere, Finland
| | - R. Puhakka
- Faculty of Biological and Environmental Sciences, University of Helsinki, Lahti, Finland
| | - A. Sinkkonen
- Natural Resources Institute Finland, Helsinki, Finland
| |
Collapse
|
158
|
Murayama MA. Complement C3 deficient mice show more severe imiquimod-induced psoriasiform dermatitis than wild-type mice regardless of the commensal microbiota. Exp Anim 2024; 73:458-467. [PMID: 38945882 PMCID: PMC11534491 DOI: 10.1538/expanim.24-0043] [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: 04/23/2024] [Accepted: 06/26/2024] [Indexed: 07/02/2024] Open
Abstract
The complement active product, C3a, and the receptor C3aR comprise an axis that exerts various biological functions, such as protection against infection. C3a is highly expressed in the inflamed skin and blood from patients with psoriasiform dermatitis. However, the role of the C3a/C3aR axis in psoriasiform dermatitis remains unclear because conflicting results using C3-/- mice have been published. In this study, to elucidate the contribution of commensal microbiota in C3-/- and wild-type (WT) mice were subjected to imiquimod-induced psoriasiform dermatitis under different housing conditions. C3-/- mice showed increased epidermal thickness and keratinocyte proliferation markers in the inflamed ear compared to WT mice upon treatment with IMQ. These inflamed phenotypes were observed in both cohoused and separately housed conditions, and antibiotic treatment did not abolish the aggravation of IMQ-induced psoriasiform dermatitis in C3-/- mice. These results suggested that the difference of commensal microbiota is not important for the C3-involved psoriasiform dermatitis. Keratinocyte hyperproliferation is a major feature of the inflamed skin in patients with psoriasiform dermatitis. In vitro experiments showed that C3a and C3aR agonists inhibited keratinocyte proliferation, which was abolished by introduction of a C3aR antagonist. Collectively, these results suggest that the C3a/C3aR axis plays a critical role in psoriasiform dermatitis development by inhibiting keratinocyte proliferation, regardless of the regulation of the commensal microbiota.
Collapse
Affiliation(s)
- Masanori A Murayama
- Department of Animal Models for Human Diseases, Institute of Biomedical Science, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka 573-1010, Japan
| |
Collapse
|
159
|
Belnap N, Ramsey K, Carvalho ST, Nearman L, Haas H, Huentelman M, Lee K. Exploring the Frontier: The Human Microbiome's Role in Rare Childhood Neurological Diseases and Epilepsy. Brain Sci 2024; 14:1051. [PMID: 39595814 PMCID: PMC11592123 DOI: 10.3390/brainsci14111051] [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: 10/06/2024] [Revised: 10/19/2024] [Accepted: 10/19/2024] [Indexed: 11/28/2024] Open
Abstract
Emerging research into the human microbiome, an intricate ecosystem of microorganisms residing in and on our bodies, reveals that it plays a pivotal role in maintaining our health, highlighting the potential for microbiome-based interventions to prevent, diagnose, treat, and manage a myriad of diseases. The objective of this review is to highlight the importance of microbiome studies in enhancing our understanding of rare genetic epilepsy and related neurological disorders. Studies suggest that the gut microbiome, acting through the gut-brain axis, impacts the development and severity of epileptic conditions in children. Disruptions in microbial composition can affect neurotransmitter systems, inflammatory responses, and immune regulation, which are all critical factors in the pathogenesis of epilepsy. This growing body of evidence points to the potential of microbiome-targeted therapies, such as probiotics or dietary modifications, as innovative approaches to managing epilepsy. By harnessing the power of the microbiome, we stand to develop more effective and personalized treatment strategies for children affected by this disease and other rare neurological diseases.
Collapse
Affiliation(s)
- Newell Belnap
- Center for Rare Childhood Disorders, Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (N.B.)
| | - Keri Ramsey
- Center for Rare Childhood Disorders, Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (N.B.)
| | | | - Lexi Nearman
- Department of Biological Science, Northern Arizona University, Flagstaff, AZ 86011, USA
- TGen Integrated Microbiomics Center, Translational Genomics Research Institute (TGen), Flagstaff, AZ 86011, USA
| | - Hannah Haas
- Center for Rare Childhood Disorders, Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (N.B.)
- Barrett, the Honors College, Arizona State University, Tempe, AZ 85281, USA
| | - Matt Huentelman
- Center for Rare Childhood Disorders, Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (N.B.)
| | - Keehoon Lee
- TGen Integrated Microbiomics Center, Translational Genomics Research Institute (TGen), Flagstaff, AZ 86011, USA
| |
Collapse
|
160
|
Chaudhary S, Kaur P, Singh TA, Bano KS, Vyas A, Mishra AK, Singh P, Mehdi MM. The dynamic crosslinking between gut microbiota and inflammation during aging: reviewing the nutritional and hormetic approaches against dysbiosis and inflammaging. Biogerontology 2024; 26:1. [PMID: 39441393 DOI: 10.1007/s10522-024-10146-2] [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: 08/02/2024] [Accepted: 10/01/2024] [Indexed: 10/25/2024]
Abstract
The early-life gut microbiota (GM) is increasingly recognized for its contributions to human health and disease over time. Microbiota composition, influenced by factors like race, geography, lifestyle, and individual differences, is subject to change. The GM serves dual roles, defending against pathogens and shaping the host immune system. Disruptions in microbial composition can lead to immune dysregulation, impacting defense mechanisms. Additionally, GM aids digestion, releasing nutrients and influencing physiological systems like the liver, brain, and endocrine system through microbial metabolites. Dysbiosis disrupts intestinal homeostasis, contributing to age-related diseases. Recent studies are elucidating the bacterial species that characterize a healthy microbiota, defining what constitutes a 'healthy' colonic microbiota. The present review article focuses on the importance of microbiome composition for the development of homeostasis and the roles of GM during aging and the age-related diseases caused by the alteration in gut microbial communities. This article might also help the readers to find treatments targeting GM for the prevention of various diseases linked to it effectively.
Collapse
Affiliation(s)
- Sakshi Chaudhary
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Pardeep Kaur
- Department of Biotechnology, Chandigarh University, Mohali, Punjab, 140413, India
| | - Thokchom Arjun Singh
- Department of Microbiology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Kaniz Shahar Bano
- Department of Microbiology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Ashish Vyas
- Department of Microbiology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Alok Kumar Mishra
- Department of Microbiology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Prabhakar Singh
- Department of Biotechnology, School of Bio and Chemical Engineering, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, 600119, India
| | - Mohammad Murtaza Mehdi
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India.
| |
Collapse
|
161
|
Zhang X, Liu Z, Liu G, Wei Z, Qin Z, Li R, Liu Y, Jiang Z, Min Y, Peng X. Causal effect of gut microbiota on occurrence of herpes zoster and postherpetic neuralgia, and role of Tyzzerella 3. Eur J Med Res 2024; 29:511. [PMID: 39438941 PMCID: PMC11515789 DOI: 10.1186/s40001-024-02106-w] [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: 10/05/2023] [Accepted: 10/12/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Previous research has established connections between gut microbiota, immune modulation, and several virus-related diseases. However, no study has explored the relationships between gut microbiota and herpes zoster and postherpetic neuralgia (PHN). METHODS A total of 205 taxa of gut microbiota were regarded as exposures. The occurrences of herpes zoster and PHN were selected as outcomes. The causal effects of gut microbiota on herpes zoster and PHN were estimated with multiple methods for two-sample Mendelian randomization, such as inverse variance weighted (IVW), MR-Egger, and weighted median. All results were subjected to FDR correction to prevent from possibility of multiple comparison. RESULTS Among the significant findings, four taxa and one genus were identified as facilitators of herpes zoster and PHN, respectively. Conversely, six genera and eleven taxa were found to inhibit herpes zoster and PHN, respectively. The causal effect of the Tyzzerella 3 was confirmed through FDR correction, making it a key focus in this study. Specifically, it was found to causally facilitate herpes zoster primarily with IVW (OR 1.420, 95% CI 1.174-1.718, p < 0.001, q = 0.039), as there is no heterogeneity or horizontal pleiotropy found. CONCLUSIONS With investigation of the causal association between gut microbiota, and herpes zoster/PHN, significant findings were identified in 22 different taxa. Among them, Tyzzerella 3 keeps significant after multiple comparison correction, and displays potential to facilitate the occurrence of herpes zoster.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zheran Liu
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Guihong Liu
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zhigong Wei
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zijian Qin
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ruidan Li
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yingtong Liu
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Zheng Jiang
- Department of Otolaryngology-Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Min
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xingchen Peng
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
162
|
Juárez-Trujillo N, Alvarado-Olivarez M, Sánchez-Mendizabal S, Jiménez-Fernández M. Effect of oral administration of chlorine dioxide on hematological, physiological parameters and intestinal microbiota in a murine model. Food Chem Toxicol 2024; 194:115068. [PMID: 39447834 DOI: 10.1016/j.fct.2024.115068] [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: 07/30/2024] [Revised: 10/14/2024] [Accepted: 10/18/2024] [Indexed: 10/26/2024]
Abstract
Chlorine dioxide (CD) is a broad-spectrum disinfectant agent used to disinfect food products and, more recently, to treat diseases such as avian influenza and COVID-19 due to its potent antimicrobial properties. However, excessive use of chlorine dioxide can cause adverse health effects. While numerous in vivo studies have evaluated its toxicity CD's toxicity, few have investigated its impact on the intestinal microbiota. This study assesses the effects of oral administration of CD on hematological and physiological parameters, and the intestinal microbiota in a murine model. CD was produced at a concentration of 16578.62 mg/L after 7 days of reaction. Results showed a significant antimicrobial effect on dietary yeasts compared to probiotics. There were significant changes in the percentages of Firmicutes (CD 81.9-87.1%, control 63.05%) and Bacteroidetes (DC 3.4-4.5%, control 22.5%) in the CD administered groups compared to the control group. CD exposure showed toxicity in hematological parameters. Additionally, consuming CD for 90 days at 10 mg/kg body weight caused colon and cecum damage and decreased the rats' weight. These findings indicate than even low doses of CD can negative effect on the microbiota, the morphology of the cecum and colon, and body weight, suggesting that prolonged consumption should be avoided.
Collapse
Affiliation(s)
- Naida Juárez-Trujillo
- Centro de Investigación y Desarrollo en Alimentos, Universidad Veracruzana, Xalapa, Veracruz, Mexico
| | | | - Salvador Sánchez-Mendizabal
- Doctorado en Investigaciones Cerebrales, Instituto de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa, Veracruz, Mexico
| | - Maribel Jiménez-Fernández
- Centro de Investigación y Desarrollo en Alimentos, Universidad Veracruzana, Xalapa, Veracruz, Mexico.
| |
Collapse
|
163
|
Do NM. From Leaky Gut to Leaky Skin: A Clinical Review of Lifestyle Influences on the Microbiome. Am J Lifestyle Med 2024:15598276241292605. [PMID: 39540169 PMCID: PMC11556587 DOI: 10.1177/15598276241292605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
The microbiome connects the gut health to the rest of the body's organs, including the skin. The pathophysiology of intestinal dysbiosis leads to the expression "leaky gut." Ongoing speculations are aimed at restoring the gut microbiota via modulating lifestyle habits to prevent and potentially reverse autoimmunity. This review finds the connections between gut dysbiosis and skin conditions. It also collects evidence of some lifestyle pillars that influence the gut microbiome including dietary intakes, exercise, sleep, stress, and toxin exposures. It addresses why maintaining a healthy intestinal microbiome is important for the health of all organs in the human host. More people are affected by gut dysbiosis resulting in pro-inflammatory effects on different organs, like the cutaneous tissue, one of the largest epithelial surfaces. It is essential to take care of the gut health because this is where most of the immune system resides. The connection between the intestinal tract with its microbiota and the cutaneous system with its microbiota seems to be mediated by the immune system of the human host. Therefore, this review enhances the understanding of the research on the gut microbiome, its relationship to skin health, and the interplay between the gut and various autoimmune cutaneous conditions.
Collapse
Affiliation(s)
- Nhi Ma Do
- Arizona School of Health Sciences, A.T. Still University, Ormond Beach, FL, USA (NMD)
| |
Collapse
|
164
|
Mousa WK, Al Ali A. The Gut Microbiome Advances Precision Medicine and Diagnostics for Inflammatory Bowel Diseases. Int J Mol Sci 2024; 25:11259. [PMID: 39457040 PMCID: PMC11508888 DOI: 10.3390/ijms252011259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/12/2024] [Accepted: 10/13/2024] [Indexed: 10/28/2024] Open
Abstract
The gut microbiome emerges as an integral component of precision medicine because of its signature variability among individuals and its plasticity, which enables personalized therapeutic interventions, especially when integrated with other multiomics data. This promise is further fueled by advances in next-generation sequencing and metabolomics, which allow in-depth high-precision profiling of microbiome communities, their genetic contents, and secreted chemistry. This knowledge has advanced our understanding of our microbial partners, their interaction with cellular targets, and their implication in human conditions such as inflammatory bowel disease (IBD). This explosion of microbiome data inspired the development of next-generation therapeutics for treating IBD that depend on manipulating the gut microbiome by diet modulation or using live products as therapeutics. The current landscape of artificial microbiome therapeutics is not limited to probiotics and fecal transplants but has expanded to include community consortia, engineered probiotics, and defined metabolites, bypassing several limitations that hindered rapid progress in this field such as safety and regulatory issues. More integrated research will reveal new therapeutic targets such as enzymes or receptors mediating interactions between microbiota-secreted molecules that drive or modulate diseases. With the shift toward precision medicine and the enhanced integration of host genetics and polymorphism in treatment regimes, the following key questions emerge: How can we effectively implement microbiomics to further personalize the treatment of diseases like IBD, leveraging proven and validated microbiome links? Can we modulate the microbiome to manage IBD by altering the host immune response? In this review, we discuss recent advances in understanding the mechanism underpinning the role of gut microbes in driving or preventing IBD. We highlight developed targeted approaches to reverse dysbiosis through precision editing of the microbiome. We analyze limitations and opportunities while defining the specific clinical niche for this innovative therapeutic modality for the treatment, prevention, and diagnosis of IBD and its potential implication in precision medicine.
Collapse
Affiliation(s)
- Walaa K. Mousa
- College of Pharmacy, Al Ain University of Science and Technology, Abu Dhabi 64141, United Arab Emirates;
- College of Pharmacy, Mansoura University, Mansoura 35516, Egypt
- AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi 112612, United Arab Emirates
| | - Aya Al Ali
- College of Pharmacy, Al Ain University of Science and Technology, Abu Dhabi 64141, United Arab Emirates;
- AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi 112612, United Arab Emirates
| |
Collapse
|
165
|
Patiyal S, Dhall A, Kumar N, Raghava GPS. HLA-DR4Pred2: An improved method for predicting HLA-DRB1*04:01 binders. Methods 2024; 232:18-28. [PMID: 39433152 DOI: 10.1016/j.ymeth.2024.10.007] [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: 07/22/2024] [Revised: 09/27/2024] [Accepted: 10/15/2024] [Indexed: 10/23/2024] Open
Abstract
HLA-DRB1*04:01 is associated with numerous diseases, including sclerosis, arthritis, diabetes, and COVID-19, emphasizing the need to scan for binders in the antigens to develop immunotherapies and vaccines. Current prediction methods are often limited by their reliance on the small datasets. This study presents HLA-DR4Pred2, developed on a large dataset containing 12,676 binders and an equal number of non-binders. It's an improved version of HLA-DR4Pred, which was trained on a small dataset, containing 576 binders and an equal number of non-binders. All models were trained, optimized, and tested on 80 % of the data using five-fold cross-validation and evaluated on the remaining 20 %. A range of machine learning techniques was employed, achieving maximum AUROC of 0.90 and 0.87, using composition and binary profile features, respectively. The performance of the composition-based model increased to 0.93, when combined with BLAST search. Additionally, models developed on the realistic dataset containing 12,676 binders and 86,300 non-binders, achieved a maximum AUROC of 0.99. Our proposed method outperformed existing methods when we compared the performance of our best model to that of existing methods on the independent dataset. Finally, we developed a standalone tool and a webserver for HLADR4Pred2, enabling the prediction, design, and virtual scanning of HLA-DRB1*04:01 binding peptides, and we also released a Python package available on the Python Package Index (https://webs.iiitd.edu.in/raghava/hladr4pred2/; https://github.com/raghavagps/hladr4pred2; https://pypi.org/project/hladr4pred2/).
Collapse
Affiliation(s)
- Sumeet Patiyal
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Phase 3, New Delhi 110020, India.
| | - Anjali Dhall
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Phase 3, New Delhi 110020, India.
| | - Nishant Kumar
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Phase 3, New Delhi 110020, India.
| | - Gajendra P S Raghava
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Phase 3, New Delhi 110020, India.
| |
Collapse
|
166
|
Bruno A, Arnoldi I, Barzaghi B, Boffi M, Casiraghi M, Colombo B, Di Gennaro P, Epis S, Facciotti F, Ferrari N, Fesce E, Ficetola GF, Fumagalli S, Galimberti A, Ghisleni G, Nissim WG, Mainardi L, Manenti R, Messina V, Negri A, Palm E, Piga BEA, Rainisio N, Tommasi N, Labra M. The One Health approach in urban ecosystem rehabilitation: An evidence-based framework for designing sustainable cities. iScience 2024; 27:110959. [PMID: 39391715 PMCID: PMC11466616 DOI: 10.1016/j.isci.2024.110959] [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] [Indexed: 10/12/2024] Open
Abstract
Rapid urbanization has led to negative, and sometimes unintended, consequences on biodiversity and human health. While cities offer numerous advantages in meeting the basic needs of a growing population, they also pose less apparent and longer-term health costs. To address the multifaceted impacts of urbanization, an evidence-based design framework for establishing mitigation and regeneration actions is essential. Via a "One Health" approach, this perspective provides recommendations and strategies for the urban ecosystem rehabilitation of future cities, placing biodiversity and ecosystem services at the core of designing healthy and sustainable urban spaces. The framework we propose is based on a Hub and Spoke model to integrate diverse perspectives from public and private sectors and declined in a six-building-blocks structure. This will ensure that efforts are sustainable, health-centered, socially inclusive, and grounded in high-quality data, reinforcing the essential connection between healthy environments and thriving communities.
Collapse
Affiliation(s)
- Antonia Bruno
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, piazza della Scienza, 2, 20126 Milan, Italy
| | - Irene Arnoldi
- Department of Biosciences, University of Milan, via Celoria, 26, 20133 Milan, Italy
| | - Benedetta Barzaghi
- Department of Environmental Science and Policy, University of Milan, via Celoria, 2, 20133 Milan, Italy
| | - Marco Boffi
- Department of Cultural Heritage and Environment, University of Milan, via Noto, 6, 20142 Milan, Italy
| | - Maurizio Casiraghi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, piazza della Scienza, 2, 20126 Milan, Italy
| | - Beatrice Colombo
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, piazza della Scienza, 2, 20126 Milan, Italy
| | - Patrizia Di Gennaro
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, piazza della Scienza, 2, 20126 Milan, Italy
| | - Sara Epis
- Department of Biosciences, University of Milan, via Celoria, 26, 20133 Milan, Italy
| | - Federica Facciotti
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, piazza della Scienza, 2, 20126 Milan, Italy
| | - Nicola Ferrari
- Department of Veterinary Medicine and Animal Sciences (DiVAS), University of Milan, via dell’Università, 6, 26900 Lodi, Italy
| | - Elisa Fesce
- Department of Veterinary Medicine and Animal Sciences (DiVAS), University of Milan, via dell’Università, 6, 26900 Lodi, Italy
| | | | - Sara Fumagalli
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, piazza della Scienza, 2, 20126 Milan, Italy
| | - Andrea Galimberti
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, piazza della Scienza, 2, 20126 Milan, Italy
| | - Giulia Ghisleni
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, piazza della Scienza, 2, 20126 Milan, Italy
| | - Werther Guidi Nissim
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, piazza della Scienza, 2, 20126 Milan, Italy
| | - Luca Mainardi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, via Ponzio, 34, 20133 Milan, Italy
| | - Raoul Manenti
- Department of Environmental Science and Policy, University of Milan, via Celoria, 2, 20133 Milan, Italy
| | - Valeria Messina
- Department of Environmental Science and Policy, University of Milan, via Celoria, 2, 20133 Milan, Italy
| | - Agata Negri
- Department of Biosciences, University of Milan, via Celoria, 26, 20133 Milan, Italy
| | - Emily Palm
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, piazza della Scienza, 2, 20126 Milan, Italy
| | - Barbara Ester Adele Piga
- Department of Architecture and Urban Studies, Laboratorio di Simulazione Urbana Fausto Curti, Politecnico di Milano, piazza da Vinci, 26, 20133 Milan, Italy
| | - Nicola Rainisio
- Department of Cultural Heritage and Environment, University of Milan, via Noto, 6, 20142 Milan, Italy
| | - Nicola Tommasi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, piazza della Scienza, 2, 20126 Milan, Italy
| | - Massimo Labra
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, piazza della Scienza, 2, 20126 Milan, Italy
| |
Collapse
|
167
|
Tang J, Lei Y, Pignalosa A, Hsu HH, Abdul-Sater AA, Sweeney G. Development of a non-invasive bioassay for adiponectin target engagement in mice. iScience 2024; 27:110994. [PMID: 39435143 PMCID: PMC11492082 DOI: 10.1016/j.isci.2024.110994] [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: 06/11/2024] [Revised: 07/23/2024] [Accepted: 09/16/2024] [Indexed: 10/23/2024] Open
Abstract
Adiponectin-based therapeutic strategies are promising for managing metabolic diseases and reducing inflammation, prompting the development of adiponectin receptor agonists. However, monitoring their pharmacodynamic actions in clinical applications is challenging. This study aimed to identify peripheral biomarkers to monitor adiponectin actions using ALY688, an adiponectin receptor agonist peptide. RNA sequencing analysis of whole blood identified a cluster of genes that were significantly increased in the ALY688-treated group compared to the control. This gene cluster was validated by qPCR and further confirmed in human peripheral blood mononuclear cells treated with ALY688 ex vivo. We also confirmed a functional outcome of ALY688 action in mice as our study also demonstrated the anti-inflammatory effect of ALY688 in a sublethal LPS mouse model. In summary, a newly identified gene cluster signature is suitable for assessing the pharmacodynamic action of adiponectin or its mimetics in blood samples.
Collapse
Affiliation(s)
- Jialing Tang
- Department of Biology, York University, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
| | - Yubin Lei
- Department of Biology, York University, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
| | - Angelica Pignalosa
- Allysta Pharmaceuticals Inc., 500 108th Avenue NE, Suite 1100, Bellevue, WA 98004, USA
| | - Henry H. Hsu
- Allysta Pharmaceuticals Inc., 500 108th Avenue NE, Suite 1100, Bellevue, WA 98004, USA
| | - Ali A. Abdul-Sater
- School of Kinesiology and Health Science, York University, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
| | - Gary Sweeney
- Department of Biology, York University, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
| |
Collapse
|
168
|
Ilan Y. Using the Constrained Disorder Principle to Navigate Uncertainties in Biology and Medicine: Refining Fuzzy Algorithms. BIOLOGY 2024; 13:830. [PMID: 39452139 PMCID: PMC11505099 DOI: 10.3390/biology13100830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 09/17/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024]
Abstract
Uncertainty in biology refers to situations in which information is imperfect or unknown. Variability, on the other hand, is measured by the frequency distribution of observed data. Biological variability adds to the uncertainty. The Constrained Disorder Principle (CDP) defines all systems in the universe by their inherent variability. According to the CDP, systems exhibit a degree of variability necessary for their proper function, allowing them to adapt to changes in their environments. Per the CDP, while variability differs from uncertainty, it can be viewed as a regulated mechanism for efficient functionality rather than uncertainty. This paper explores the various aspects of un-certainties in biology. It focuses on using CDP-based platforms for refining fuzzy algorithms to address some of the challenges associated with biological and medical uncertainties. Developing a fuzzy decision tree that considers the natural variability of systems can help minimize uncertainty. This method can reveal previously unidentified classes, reduce the number of unknowns, improve the accuracy of modeling results, and generate algorithm outputs that are more biologically and clinically relevant.
Collapse
Affiliation(s)
- Yaron Ilan
- Department of Medicine, Hadassah Medical Center, Faculty of Medicine, Hebrew University, Jerusalem 9112001, Israel
| |
Collapse
|
169
|
Delgado-Ocaña S, Cuesta S. From microbes to mind: germ-free models in neuropsychiatric research. mBio 2024; 15:e0207524. [PMID: 39207144 PMCID: PMC11481874 DOI: 10.1128/mbio.02075-24] [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] [Indexed: 09/04/2024] Open
Abstract
The gut-microbiota-brain axis refers to the bidirectional communication system between the gut, its microbial community, and the brain. This interaction involves a complex interplay of neural pathways, chemical transmitters, and immunological mechanisms. Germ-free animal models have been extensively employed to investigate gut-microbiota-brain interactions, significantly contributing to our current understanding of the role of intestinal microbes in brain function. However, despite the many benefits, this absence of microbiota is not futile. Germ-free animals present physiological and neurodevelopmental alterations that can persist even after reconstitution with normal microbiota. Therefore, the main goal of this minireview is to discuss how some of the inherent limitations of this model can interfere with the conclusion obtained when using these animals to study the complex nature of neuropsychiatric disorders. Furthermore, we examine the inclusion and use of antibiotic-based treatments as an alternative in the research of gut-brain interactions.
Collapse
Affiliation(s)
- Susana Delgado-Ocaña
- Department of Cell Biology and Neuroscience, Rutgers the State University of New Jersey, Piscataway, New Jersey, USA
| | - Santiago Cuesta
- Department of Cell Biology and Neuroscience, Rutgers the State University of New Jersey, Piscataway, New Jersey, USA
| |
Collapse
|
170
|
Wrona MV, Ghosh R, Coll K, Chun C, Yousefzadeh MJ. The 3 I's of immunity and aging: immunosenescence, inflammaging, and immune resilience. FRONTIERS IN AGING 2024; 5:1490302. [PMID: 39478807 PMCID: PMC11521913 DOI: 10.3389/fragi.2024.1490302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024]
Abstract
As we age, our immune system's ability to effectively respond to pathogens declines, a phenomenon known as immunosenescence. This age-related deterioration affects both innate and adaptive immunity, compromising immune function and leading to chronic inflammation that accelerates aging. Immunosenescence is characterized by alterations in immune cell populations and impaired functionality, resulting in increased susceptibility to infections, diminished vaccine efficacy, and higher prevalence of age-related diseases. Chronic low-grade inflammation further exacerbates these issues, contributing to a decline in overall health and resilience. This review delves into the characteristics of immunosenescence and examines the various intrinsic and extrinsic factors contributing to immune aging and how the hallmarks of aging and cell fates can play a crucial role in this process. Additionally, it discusses the impact of sex, age, social determinants, and gut microbiota health on immune aging, illustrating the complex interplay of these factors in altering immune function. Furthermore, the concept of immune resilience is explored, focusing on the metrics for assessing immune health and identifying strategies to enhance immune function. These strategies include lifestyle interventions such as diet, regular physical activity, stress management, and the use of gerotherapeutics and other approaches. Understanding and mitigating the effects of immunosenescence are crucial for developing interventions that support robust immune responses in aged individuals.
Collapse
Affiliation(s)
- Marianna V. Wrona
- Columbia University in the City of New York, New York, NY, United States
| | - Rituparna Ghosh
- Columbia Center for Human Longevity, Columbia University Medical Center, New York, NY, United States
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, United States
- Department of Medicine, Columbia University Medical Center, New York, NY, United States
| | - Kaitlyn Coll
- Florida International University, Miami, FL, United States
| | - Connor Chun
- Bronx High School of Science, New York, NY, United States
| | - Matthew J. Yousefzadeh
- Columbia University in the City of New York, New York, NY, United States
- Columbia Center for Human Longevity, Columbia University Medical Center, New York, NY, United States
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, United States
- Department of Medicine, Columbia University Medical Center, New York, NY, United States
| |
Collapse
|
171
|
Reddiar SB, Xie Y, Abdallah M, Han S, Hu L, Feeney OM, Gracia G, Anshabo A, Lu Z, Farooq MA, Styles IK, Phillips ARJ, Windsor JA, Porter CJH, Cao E, Trevaskis NL. Intestinal Lymphatic Biology, Drug Delivery, and Therapeutics: Current Status and Future Directions. Pharmacol Rev 2024; 76:1326-1398. [PMID: 39179383 DOI: 10.1124/pharmrev.123.001159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 07/29/2024] [Accepted: 08/14/2024] [Indexed: 08/26/2024] Open
Abstract
Historically, the intestinal lymphatics were considered passive conduits for fluids, immune cells, dietary lipids, lipid soluble vitamins, and lipophilic drugs. Studies of intestinal lymphatic drug delivery in the late 20th century focused primarily on the drugs' physicochemical properties, especially high lipophilicity, that resulted in intestinal lymphatic transport. More recent discoveries have changed our traditional view by demonstrating that the lymphatics are active, plastic, and tissue-specific players in a range of biological and pathological processes, including within the intestine. These findings have, in turn, inspired exploration of lymph-specific therapies for a range of diseases, as well as the development of more sophisticated strategies to actively deliver drugs or vaccines to the intestinal lymph, including a range of nanotechnologies, lipid prodrugs, and lipid-conjugated materials that "hitchhike" onto lymphatic transport pathways. With the increasing development of novel therapeutics such as biologics, there has been interest in whether these therapeutics are absorbed and transported through intestinal lymph after oral administration. Here we review the current state of understanding of the anatomy and physiology of the gastrointestinal lymphatic system in health and disease, with a focus on aspects relevant to drug delivery. We summarize the current state-of-the-art approaches to deliver drugs and quantify their uptake into the intestinal lymphatic system. Finally, and excitingly, we discuss recent examples of significant pharmacokinetic and therapeutic benefits achieved via intestinal lymphatic drug delivery. We also propose approaches to advance the development and clinical application of intestinal lymphatic delivery strategies in the future. SIGNIFICANCE STATEMENT: This comprehensive review details the understanding of the anatomy and physiology of the intestinal lymphatic system in health and disease, with a focus on aspects relevant to drug delivery. It highlights current state-of-the-art approaches to deliver drugs to the intestinal lymphatics and the shift toward the use of these strategies to achieve pharmacokinetic and therapeutic benefits for patients.
Collapse
Affiliation(s)
- Sanjeevini Babu Reddiar
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Yining Xie
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Mohammad Abdallah
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Sifei Han
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Luojuan Hu
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Orlagh M Feeney
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Gracia Gracia
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Abel Anshabo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Zijun Lu
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Muhammad Asim Farooq
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Ian K Styles
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Anthony R J Phillips
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - John A Windsor
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Christopher J H Porter
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Enyuan Cao
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Natalie L Trevaskis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| |
Collapse
|
172
|
Fathima A, Jamma T. UDCA ameliorates inflammation driven EMT by inducing TGR5 dependent SOCS1 expression in mouse macrophages. Sci Rep 2024; 14:24285. [PMID: 39414916 PMCID: PMC11484976 DOI: 10.1038/s41598-024-75516-9] [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: 05/29/2024] [Accepted: 10/07/2024] [Indexed: 10/18/2024] Open
Abstract
Long-standing chronic inflammation of the digestive tract leads to Inflammatory Bowel Diseases (IBD), comprising Crohn's Disease (CD) and Ulcerative colitis (UC). The persistent prevalence of these conditions in the gut is a predisposing factor for Colitis-Associated Cancer (CAC), one of the most common sub-types of Colorectal Cancer (CRC), emphasizing the role of inflammation in tumorigenesis. Therefore, targeted intervention of chronic intestinal inflammation is a potential strategy for preclusion and treatment of inflammation-driven malignancies. The association between bile acids (BA) and gut immune homeostasis has been explored in the recent past. However, the exact downstream mechanism by which secondary BA successfully regulating intestinal inflammation and inflammation-dependent CAC is unclear. Our study demonstrated that Ursodeoxycholic acid (UDCA), a secondary bile acid of host gut microbial origin, finetunes the dialogue between activated macrophages and intestinal epithelial cells, modulating inflammation-driven epithelial-mesenchymal transition (EMT), a hallmark of cancer. UDCA treatment and dependency on the TGR5/GPBAR1 receptor significantly upregulated the Suppressor of Cytokine Signaling 1 (SOCS1) expression, contributing to the regulation of pro-inflammatory cytokines in activated macrophages. In this study, we also noticed heightened expression of SOCS1 in UDCA-mitigated CAC in the AOM-DSS mouse model with reduced inflammatory gene expression. Overall, our observations highlight the possible utility of UDCA for inflammation-driven intestinal cancer.
Collapse
Affiliation(s)
- Ashna Fathima
- Cell Signaling Laboratory, Department of Biological Sciences, Birla Institute of Technology, and Science-Pilani Hyderabad Campus, Jawahar Nagar, Shameerpet Mandal, Hyderabad, 500078, Telangana , India
| | - Trinath Jamma
- Cell Signaling Laboratory, Department of Biological Sciences, Birla Institute of Technology, and Science-Pilani Hyderabad Campus, Jawahar Nagar, Shameerpet Mandal, Hyderabad, 500078, Telangana , India.
| |
Collapse
|
173
|
Leibovitzh H, Sarbagili Shabat C, Hirsch A, Zittan E, Mentella MC, Petito V, Cohen NA, Ron Y, Fliss Isakov N, Pfeffer J, Yaakov M, Fanali C, Turchini L, Masucci L, Quaranta G, Kolonimos N, Godneva A, Weinberger A, Scaldaferri F, Maharshak N. Faecal Transplantation for Ulcerative Colitis From Diet Conditioned Donors Followed by Dietary Intervention Results in Favourable Gut Microbial Profile Compared to Faecal Transplantation Alone. J Crohns Colitis 2024; 18:1606-1614. [PMID: 38720628 DOI: 10.1093/ecco-jcc/jjae062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/17/2024] [Accepted: 05/08/2024] [Indexed: 10/17/2024]
Abstract
BACKGROUND AND AIMS Several faecal microbial transplantation [FMT] approaches for ulcerative colitis [UC] have been investigated with conflicting results. We have recently published the clinical outcomes from the CRAFT UC Trial using FMT with the UC Exclusion Diet [UCED], compared with FMT alone. Here we aimed to compare the two FMT strategies in terms of microbial profile and function. METHODS Subjects recruited to the CRAFT UC study with available pre- and post-intervention faecal samples were included. Donors received diet conditioning for 14 days based on the UCED principles. Group 1 received single FMT by colonoscopy [Day 1] and enemas [Days 2 and 14] without donors' dietary conditioning [N = 11]. Group 2 received FMT but with donors' dietary pre-conditioning and UCED for the patients [N = 10]. Faecal samples were assessed by DNA shotgun metagenomic sequencing. RESULTS Following diet conditioning, donors showed depletion in metabolic pathways involved in biosynthesis of sulphur-containing amino acids. Only Group 2 showed significant shifts towards the donors' microbial composition [ADONIS: R2 = 0.15, p = 0.008] and significantly increased Eubacterium_sp_AF228LB post-intervention [β-coefficient 2.66, 95% confidence interval 2.1-3.3, q < 0.05] which was inversely correlated with faecal calprotectin [rho = -0.52, p = 0.035]. Moreover, pathways involved in gut inflammation and barrier function including branched chain amino acids were enriched post-intervention in Group 2 and were significantly inversely correlated with faecal calprotectin. CONCLUSION FMT from diet conditioned donors followed by the UCED led to microbial alterations associated with favourable microbial profiles which correlated with decreased faecal calprotectin. Our findings support further exploration of the additive benefit of dietary intervention for both donors and patients undergoing FMT as a potential treatment of UC.
Collapse
Affiliation(s)
- Haim Leibovitzh
- Department of Gastroenterology and Hepatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Chen Sarbagili Shabat
- Pediatric Gastroenterology Unit, PIBD Research Center, Wolfson Medical Center, Holon, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ayal Hirsch
- Department of Gastroenterology and Hepatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Eran Zittan
- Gastroenterology Institute, IBD Unit, Haemek Medical Center, Afula, Israel
| | - Maria Chiara Mentella
- UOC di Nutrizione Clinica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Valentina Petito
- Cemad [CENTER for Digestive Disease], UOC Medicina Internae Gastroenterologia, Fondazione Policlinico 'A. Gemelli' IRCCS, Rome, Italy
| | - Nathaniel Aviv Cohen
- Department of Gastroenterology and Hepatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Yulia Ron
- Department of Gastroenterology and Hepatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Naomi Fliss Isakov
- Department of Gastroenterology and Hepatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Department of Health, School of Public Health, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jorge Pfeffer
- Department of Gastroenterology and Hepatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Michal Yaakov
- Pediatric Gastroenterology Unit, PIBD Research Center, Wolfson Medical Center, Holon, Israel
| | - Caterina Fanali
- Cemad [CENTER for Digestive Disease], UOC Medicina Internae Gastroenterologia, Fondazione Policlinico 'A. Gemelli' IRCCS, Rome, Italy
| | - Laura Turchini
- Cemad [CENTER for Digestive Disease], UOC Medicina Internae Gastroenterologia, Fondazione Policlinico 'A. Gemelli' IRCCS, Rome, Italy
| | - Luca Masucci
- Istituto di Microbiologia, Università Cattolica del Sacro Cuore - Fondazione Policlinico 'A. Gemelli' IRCSS, Rome, Italy
- Dipartimento Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Gianluca Quaranta
- Istituto di Microbiologia, Università Cattolica del Sacro Cuore - Fondazione Policlinico 'A. Gemelli' IRCSS, Rome, Italy
- Dipartimento Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Nitzan Kolonimos
- Gastroenterology Institute, IBD Unit, Haemek Medical Center, Afula, Israel
| | - Anastasia Godneva
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel
| | - Adina Weinberger
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel
| | - Franco Scaldaferri
- Cemad [CENTER for Digestive Disease], UOC Medicina Internae Gastroenterologia, Fondazione Policlinico 'A. Gemelli' IRCCS, Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore- Fondazione Policlinico 'A. Gemelli' IRCCS, Rome, Italy
| | - Nitsan Maharshak
- Department of Gastroenterology and Hepatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
174
|
Fachi JL, Davoli Ferreira M, Câmara NOS. Editorial: Molecular insights of microbiota and innate lymphoid cell interactions. Front Immunol 2024; 15:1497305. [PMID: 39464887 PMCID: PMC11502386 DOI: 10.3389/fimmu.2024.1497305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 09/30/2024] [Indexed: 10/29/2024] Open
Affiliation(s)
- José Luís Fachi
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, United States
| | - Marcela Davoli Ferreira
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | | |
Collapse
|
175
|
Chen D, Woo JMP, Parks CG, Lawrence KG, O'Brien KM, Sandler RS, Sandler DP. Childhood and adolescent residential and farm pesticide exposures and inflammatory bowel disease incidence in a U.S. cohort of women. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 946:174475. [PMID: 38964382 PMCID: PMC11296211 DOI: 10.1016/j.scitotenv.2024.174475] [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: 05/06/2024] [Revised: 07/01/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND AND AIMS There are few known risk factors for inflammatory bowel disease (IBD), an autoimmune disease characterized by chronic intestinal inflammation. Use of specific pesticides has been associated with higher incidence of IBD among pesticide applicators and their spouses, but no study has examined pesticide exposure in early life, a period where the human immune system undergoes rapid changes. We evaluated pesticide use during childhood and adolescence and incidence of IBD among US women enrolled in the Sister Study. METHODS Incident IBD diagnoses between enrollment (2003-2009) and 2021 were identified and validated with medication use and colectomy/colostomy surgery. We estimated hazard ratios (HR) and 95 % confidence intervals (CI) for the relationship of childhood/adolescent residential and farm pesticide exposures with IBD incidence using Cox models, accounting for age, race and ethnicity, education, smoking, and birth year. RESULTS We identified 277 incident IBD cases among 48,382 eligible participants. IBD hazard was elevated among those whose childhood residence was regularly treated with pesticides, especially among those who ever personally applied pesticides (HR = 1.39, 95%CI: 0.65, 2.99). We observed a positive association between IBD and exposure to broadcast pesticide sprays before DDT was banned (>6 times vs. never HR = 1.56, 95%CI: 1.06, 2.31). Among participants who lived on a farm during childhood/adolescence for ≥1 year (N = 9162), IBD hazards were higher among those who were in crop fields during pesticide application (HR = 2.06, 95%CI: 0.94, 4.51) and who ever personally applied pesticides on crops (HR = 1.85, 95%CI: 0.81, 4.18) or livestock (HR = 2.58, 95%CI: 1.14, 5.83). CONCLUSION Early-life pesticide exposure may be a novel risk factor for IBD. Practices that reduce pesticide exposure during early life may help reduce the burden of this disease.
Collapse
Affiliation(s)
- Dazhe Chen
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Jennifer M P Woo
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Christine G Parks
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Kaitlyn G Lawrence
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Katie M O'Brien
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Robert S Sandler
- Division of Gastroenterology and Hepatology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Dale P Sandler
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA.
| |
Collapse
|
176
|
Sandhanam K, Tamilanban T, Bhattacharjee B, Manasa K. Exploring miRNA therapies and gut microbiome-enhanced CAR-T cells: advancing frontiers in glioblastoma stem cell targeting. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03479-9. [PMID: 39382681 DOI: 10.1007/s00210-024-03479-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 09/20/2024] [Indexed: 10/10/2024]
Abstract
Glioblastoma multiforme (GBM) presents a formidable challenge in oncology due to its aggressive nature and resistance to conventional treatments. Recent advancements propose a novel therapeutic strategy combining microRNA-based therapies, chimeric antigen receptor-T (CAR-T) cells, and gut microbiome modulation to target GBM stem cells and transform cancer treatment. MicroRNA therapies show promise in regulating key signalling pathways implicated in GBM progression, offering the potential to disrupt GBM stem cell renewal. CAR-T cell therapy, initially successful in blood cancers, is being adapted to target GBM by genetically engineering T cells to recognise and eliminate GBM stem cell-specific antigens. Despite early successes, challenges like the immunosuppressive tumour microenvironment persist. Additionally, recent research has uncovered a link between the gut microbiome and GBM, suggesting that gut dysbiosis can influence systemic inflammation and immune responses. Novel strategies to modulate the gut microbiome are emerging, enhancing the efficacy of microRNA therapies and CAR-T cell treatments. This combined approach highlights the synergistic potential of these innovative therapies in GBM treatment, aiming to eradicate primary tumours and prevent recurrence, thereby improving patient prognosis and quality of life. Ongoing research and clinical trials are crucial to fully exploit this promising frontier in GBM therapy, offering hope to patients grappling with this devastating disease.
Collapse
Affiliation(s)
- K Sandhanam
- Department of Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology, Chengalpattu, 603203, Tamil Nadu, India
| | - T Tamilanban
- Department of Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology, Chengalpattu, 603203, Tamil Nadu, India.
| | - Bedanta Bhattacharjee
- Department of Pharmacology, Girijananda Chowdhury University-Tezpur Campus, 784501, Assam, India
| | - K Manasa
- Department of Pharmacology, MNR College of Pharmacy, Sangareddy, 502294, Telangana, India
| |
Collapse
|
177
|
Marangelo C, Vernocchi P, Del Chierico F, Scanu M, Marsiglia R, Petrolo E, Fucà E, Guerrera S, Valeri G, Vicari S, Putignani L. Stratification of Gut Microbiota Profiling Based on Autism Neuropsychological Assessments. Microorganisms 2024; 12:2041. [PMID: 39458350 PMCID: PMC11510388 DOI: 10.3390/microorganisms12102041] [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: 09/13/2024] [Revised: 10/04/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder. Investigations of gut microbiota (GM) play an important role in deciphering disease severity and symptoms. Overall, we stratified 70 ASD patients by neuropsychological assessment, based on Calibrated Severity Scores (CSSs) of the Autism Diagnostic Observation Schedule-Second edition (ADOS-2), Child Behavior Checklist (CBCL) and intelligent quotient/developmental quotient (IQ/DQ) parameters. Hence, metataxonomy and PICRUSt-based KEGG predictions of fecal GM were assessed for each clinical subset. Here, 60% of ASD patients showed mild to moderate autism, while the remaining 40% showed severe symptoms; 23% showed no clinical symptoms, 21% had a risk of behavior problems and 56% had clinical symptoms based on the CBCL, which assesses internalizing problems; further, 52% had no clinical symptoms, 21% showed risk, and 26% had clinical symptoms classified by CBCL externalizing problems. Considering the total CBCL index, 34% showed no clinical symptoms, 13% showed risk, and 52% had clinical symptoms. Here, 70% of ASD patients showed cognitive impairment/developmental delay (CI/DD). The GM of ASDs with severe autism was characterized by an increase in Veillonella, a decrease in Monoglobus pectinilyticus and a higher microbial dysbiosis index (MDI) when compared to mild-moderate ASDs. Patients at risk for behavior problems and showing clinical symptoms were characterized by a GM with an increase of Clostridium, Eggerthella, Blautia, Intestinibacter, Coprococcus, Ruminococcus, Onthenecus and Bariatricus, respectively. Peptidoglycan biosynthesis and biofilm formation KEGGs characterized patients with clinical symptoms, while potential microbiota-activated PPAR-γ-signaling was seen in CI/DD patients. This evidence derived from GM profiling may be used to further improve ASD understanding, leasing to a better comprehension of the neurological phenotype.
Collapse
Affiliation(s)
- Chiara Marangelo
- Research Unit of Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (C.M.); (P.V.); (F.D.C.); (M.S.); (R.M.)
| | - Pamela Vernocchi
- Research Unit of Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (C.M.); (P.V.); (F.D.C.); (M.S.); (R.M.)
| | - Federica Del Chierico
- Research Unit of Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (C.M.); (P.V.); (F.D.C.); (M.S.); (R.M.)
| | - Matteo Scanu
- Research Unit of Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (C.M.); (P.V.); (F.D.C.); (M.S.); (R.M.)
| | - Riccardo Marsiglia
- Research Unit of Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (C.M.); (P.V.); (F.D.C.); (M.S.); (R.M.)
| | - Emanuela Petrolo
- Child and Adolescent Neuropsychiatry Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (E.P.); (E.F.); (S.G.); (G.V.); or (S.V.)
| | - Elisa Fucà
- Child and Adolescent Neuropsychiatry Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (E.P.); (E.F.); (S.G.); (G.V.); or (S.V.)
| | - Silvia Guerrera
- Child and Adolescent Neuropsychiatry Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (E.P.); (E.F.); (S.G.); (G.V.); or (S.V.)
| | - Giovanni Valeri
- Child and Adolescent Neuropsychiatry Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (E.P.); (E.F.); (S.G.); (G.V.); or (S.V.)
| | - Stefano Vicari
- Child and Adolescent Neuropsychiatry Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (E.P.); (E.F.); (S.G.); (G.V.); or (S.V.)
- Life Sciences and Public Health Department, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Lorenza Putignani
- Unit of Microbiomics and Research Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy
| |
Collapse
|
178
|
Ren R, Huang X, Wei D, Guo Q, Wang C, Li M, Yang L, Lang H, Chen S. Microbe-immune interactions: new perspectives on coagulation deficiencies, purpura, and other hemorrhagic conditions under the regulation of the gut microbiota. Front Immunol 2024; 15:1461221. [PMID: 39439804 PMCID: PMC11493621 DOI: 10.3389/fimmu.2024.1461221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 09/11/2024] [Indexed: 10/25/2024] Open
Abstract
Background The relationship between gut microbiota and coagulation defects, purpura, and other hemorrhagic conditions (CPH) is currently unclear, with causal links yet to be firmly established. Objective The causal relationships between gut microbiota and CPH, along with the potential mediating role of immune cells, were studied using Mendelian randomization analysis. Methods Data on 412 gut microbiota species, 731 immune cell types, and CPH were methodologically compiled from genome-wide association studies and the FinnGen database. A 2-sample Mendelian randomization approach in 2 stages was used and the causal links between gut microbiota and CPH were statistically analyzed, assessing the potential mediation by immune cells. Sensitivity and reliability were ensured through heterogeneity and pleiotropy tests. Results The abundance of Alistipes putredinis (odds ratio [OR]=0.77, 95% confidence interval [CI] 0.64-0.93, P=0.006) was negatively correlated with CPH, whereas the abundance of Bacteroides stercoris (OR=1.25, 95%CI 1.09-1.45, P=0.002) was positively correlated with the risk of CPH. There was no evidence of reverse causality or the potential mediating effects of 731 immune cell types. The abundance of Proteobacteria (OR=0.81, 95%CI 0.71-0.92, P=0.001) and Coprococcus sp. ART55/1 (OR=0.87, 95%CI 0.80-0.96, P=0.005) was negatively associated with the risk of CPH, whereas the abundance of Enterobacteriales/Enterobacteriaceae (OR=1.36, 95%CI 1.12-1.64, P=0.002) was positively correlated with the risk of CPH, with no evidence of reverse causality. Furthermore, CD38 levels on CD3-CD19 cells can serve as a mediating factor for the influence of Proteobacteria on the pathogenesis of CPH, with a mediating effect ratio of 7.26%. Conclusions An increase in Proteobacteria abundance leads to a decrease in CD38 expression on CD3-CD19- cells, thereby reducing the risk of developing CPH. CD3 expression on naive CD4+ in mature T cells serves as a mediating factor for the influence of Enterobacteriales/Enterobacteriaceae on the pathogenesis of CPH, whereas IgD CD38br AC expression on B cells serves as a mediating factor for the influence of Coprococcus sp. ART55/1 on the pathogenesis of CPH. The mediating effect is opposite to the overall trend and has a relatively small impact. No significant heterogeneity or pleiotropy was observed.
Collapse
Affiliation(s)
- Ruhua Ren
- The First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
- Department of Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaohua Huang
- The First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
- Department of Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Diu Wei
- The First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
- Department of Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Qing Guo
- Department of Hematology, International Mongolian Hospital of Inner Mongolia, Hohhot, China
| | - Chong Wang
- The First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
- Department of Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Mengjie Li
- The First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
- Department of Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Lu Yang
- The First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
- Department of Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Haiyan Lang
- The First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
- Department of Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Shana Chen
- Department of Hematology, International Mongolian Hospital of Inner Mongolia, Hohhot, China
| |
Collapse
|
179
|
Zheng X, Chen Q, Liang X, Kumar V, Loor A, Dong H, Liu C, Yang J, Zhang J. The Composition and Function of Intestinal Microbiota Were Altered in Farmed Bullfrog Tadpoles ( Aquarana catesbeiana) during Metamorphosis. Microorganisms 2024; 12:2020. [PMID: 39458329 PMCID: PMC11510581 DOI: 10.3390/microorganisms12102020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 09/29/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024] Open
Abstract
The bullfrog Aquarana catesbeiana is one of the main farmed frog species in China, with a low overall survival of farmed bullfrogs from hatching to harvest since bullfrog tadpoles are fragile during the metamorphosis period. The intestinal bacterial community can play crucial roles in animal development; however, little is known about the alteration of the gut microbial community of A. catesbeiana during metamorphosis. The present study used 16S rRNA amplicon sequencing to investigate the intestinal bacterial community in A. catesbeiana at four distinct developmental stages. Moreover, we determined the bullfrog's body morphological parameters and the intestine histology at different developmental stages. The results showed a reduction in the total length and snout-vent length of A. catesbeiana during metamorphosis. The intestinal microbial composition of A. catesbeiana exhibited variation throughout the process of metamorphosis. The terrestrial stage showed shifts in the bacterial composition compared to the aquatic stages, including a reduction in Bacteroidetes and an increase in Firmicutes. Furthermore, the presence of Prevotella, Bifidobacterium, Leucobacter, Corynebacterium, Bulleidia, Dorea, Robinsoniella, and Clostridium in A. catesbeiana metamorphosis appears to be mainly related to the host's epithelial cells' height and total body mass. The results indicated that the intestinal microbial composition changed with the bullfrog-tadpole metamorphosis. The genera of Prevotella, Bifidobacterium, Leucobacter, Corynebacterium, Bulleidia, Dorea, Robinsoniella, and Clostridium might be potential probiotics.
Collapse
Affiliation(s)
- Xiaoting Zheng
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510300, China; (X.Z.); (Q.C.); (X.L.); (H.D.); (C.L.)
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, China;
| | - Qiuyu Chen
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510300, China; (X.Z.); (Q.C.); (X.L.); (H.D.); (C.L.)
| | - Xueying Liang
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510300, China; (X.Z.); (Q.C.); (X.L.); (H.D.); (C.L.)
| | - Vikash Kumar
- Aquatic Environmental Biotechnology and Nanotechnology (AEBN) Division, ICAR–Central Inland Fisheries Research Institute (CIFRI), Barrackpore 700120, India;
| | - Alfredo Loor
- Faculty of Maritime Engineering and Marine Sciences (FIMCM), Escuela Superior Politecnica del Litoral (ESPOL), Guayaquil 09015863, Ecuador;
| | - Hongbiao Dong
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510300, China; (X.Z.); (Q.C.); (X.L.); (H.D.); (C.L.)
- Sanya Tropical Fisheries Research Institute, Sanya 572018, China
| | - Chang Liu
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510300, China; (X.Z.); (Q.C.); (X.L.); (H.D.); (C.L.)
| | - Jinlong Yang
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, China;
| | - Jiasong Zhang
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510300, China; (X.Z.); (Q.C.); (X.L.); (H.D.); (C.L.)
- Sanya Tropical Fisheries Research Institute, Sanya 572018, China
| |
Collapse
|
180
|
Kolahi Sadeghi L, Vahidian F, Eterafi M, Safarzadeh E. Gastrointestinal cancer resistance to treatment: the role of microbiota. Infect Agent Cancer 2024; 19:50. [PMID: 39369252 PMCID: PMC11453072 DOI: 10.1186/s13027-024-00605-3] [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: 06/15/2024] [Accepted: 08/20/2024] [Indexed: 10/07/2024] Open
Abstract
The most common illnesses that adversely influence human health globally are gastrointestinal malignancies. The prevalence of gastrointestinal cancers (GICs) is relatively high, and the majority of patients receive ineffective care since they are discovered at an advanced stage of the disease. A major component of the human body is thought to be the microbiota of the gastrointestinal tract and the genes that make up the microbiome. The gut microbiota includes more than 3000 diverse species and billions of microbes. Each of them has benefits and drawbacks and been demonstrated to alter anticancer medication efficacy. Treatment of GIC with the help of the gut bacteria is effective while changes in the gut microbiome which is linked to resistance immunotherapy or chemotherapy. Despite significant studies and findings in this field, more research on the interactions between microbiota and response to treatment in GIC are needed to help researchers provide more effective therapeutic strategies with fewer treatment complication. In this review, we examine the effect of the human microbiota on anti-cancer management, including chemotherapy, immunotherapy, and radiotherapy.
Collapse
Affiliation(s)
- Leila Kolahi Sadeghi
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Fatemeh Vahidian
- Faculty of Medicine, Laval University, Quebec, Canada
- Centre de Recherche de I'Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ), Quebec, Canada
| | - Majid Eterafi
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
- Students' Research Committee, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Elham Safarzadeh
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
- Department of Microbiology, Parasitology, and Immunology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
181
|
Okolie MC, Edo GI, Ainyanbhor IE, Jikah AN, Akpoghelie PO, Yousif E, Zainulabdeen K, Isoje EF, Igbuku UA, Orogu JO, Owheruo JO, Essaghah AEA, Umar H. Gut microbiota and immunity in health and diseases: a review. PROCEEDINGS OF THE INDIAN NATIONAL SCIENCE ACADEMY 2024. [DOI: 10.1007/s43538-024-00355-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 09/24/2024] [Indexed: 01/03/2025]
|
182
|
Suárez M, Solís G, Mantecón L, Gueimonde M, Arboleya S. Impacts of Different Perinatal Factors on Faecal Immune Compounds in Infants: Determination of Normal Values. Int J Mol Sci 2024; 25:10675. [PMID: 39409004 PMCID: PMC11477395 DOI: 10.3390/ijms251910675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 10/20/2024] Open
Abstract
The gut microbiota is a key and primary stimulus for the development of a host's immune system. The early establishment of the gut microbiota is affected by several perinatal factors but little is known about their influence on shaping normal immune development and, consequently, on the programming of future health. The analysis of different immune compounds is well-documented in serum samples; however, their presence in faecal samples has not been studied, and this information could be valuable in early life. In this context, the authors of this study aimed to both describe the immunological faecal profile of a cohort of one-month-old infants and describe the impact of different perinatal factors, exploring possible associations between immune compounds and gut microbiota in faecal samples. Clear differences in immune profile were observed between full-term and premature infants. Breastfeeding increases IgG2, IgG4, and IgA; in addition, male babies showed some increased Igs, among other observations. Overall, the findings of this study reinforce the hypothesis that microorganisms and immune compounds interact with each other in the early neonatal gut and that understanding these interactions in depth will help us comprehend the influence of the gut microbiota on short- and long-term infant health outcomes.
Collapse
Affiliation(s)
- Marta Suárez
- Pediatrics Service, Central University Hospital of Asturias (HUCA-SESPA), 33011 Oviedo, Spain; (M.S.); (G.S.); (L.M.)
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
| | - Gonzalo Solís
- Pediatrics Service, Central University Hospital of Asturias (HUCA-SESPA), 33011 Oviedo, Spain; (M.S.); (G.S.); (L.M.)
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
- Department of Medicine, Faculty of Medicine and Health Sciences, Universidad de Oviedo, 33011 Oviedo, Spain
- Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Developmental Origin Network (RICORS), 33011 Oviedo, Spain
| | - Laura Mantecón
- Pediatrics Service, Central University Hospital of Asturias (HUCA-SESPA), 33011 Oviedo, Spain; (M.S.); (G.S.); (L.M.)
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
| | - Miguel Gueimonde
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain
| | - Silvia Arboleya
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain
| |
Collapse
|
183
|
Luppi S, Aldegheri L, Azzalini E, Pacetti E, Barucca Sebastiani G, Fabiani C, Robino A, Comar M. Unravelling the Role of Gut and Oral Microbiota in the Pediatric Population with Type 1 Diabetes Mellitus. Int J Mol Sci 2024; 25:10611. [PMID: 39408940 PMCID: PMC11477131 DOI: 10.3390/ijms251910611] [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: 09/02/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Type 1 Diabetes Mellitus (T1DM) is a chronic autoimmune disease that results in the destruction of pancreatic β cells, leading to hyperglycaemia and the need for lifelong insulin therapy. Although genetic predisposition and environmental factors are considered key contributors to T1DM, the exact causes of the disease remain partially unclear. Recent evidence has focused on the relationship between the gut, the oral cavity, immune regulation, and systemic inflammation. In individuals with T1DM, changes in the gut and oral microbial composition are commonly observed, indicating that dysbiosis may contribute to immune dysregulation. Gut dysbiosis can influence the immune system through increased intestinal permeability, altered production of short chain fatty acids (SCFAs), and interactions with the mucosal immune system, potentially triggering the autoimmune response. Similarly, oral dysbiosis may contribute to the development of systemic inflammation and thus influence the progression of T1DM. A comprehensive understanding of these relationships is essential for the identification of biomarkers for early diagnosis and monitoring, as well as for the development of therapies aimed at restoring microbial balance. This review presents a synthesis of current research on the connection between T1DM and microbiome dysbiosis, with a focus on the gut and oral microbiomes in pediatric populations. It explores potential mechanisms by which microbial dysbiosis contributes to the pathogenesis of T1DM and examines the potential of microbiome-based therapies, including probiotics, prebiotics, synbiotics, and faecal microbiota transplantation (FMT). This complex relationship highlights the need for longitudinal studies to monitor microbiome changes over time, investigate causal relationships between specific microbial species and T1DM, and develop personalised medicine approaches.
Collapse
Affiliation(s)
- Stefania Luppi
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (S.L.); (L.A.); (M.C.)
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy; (E.A.); (E.P.)
| | - Luana Aldegheri
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (S.L.); (L.A.); (M.C.)
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy; (E.A.); (E.P.)
| | - Eros Azzalini
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy; (E.A.); (E.P.)
| | - Emanuele Pacetti
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy; (E.A.); (E.P.)
| | - Giulia Barucca Sebastiani
- Medicine of Services Department, Clinical Analysis Laboratory, Azienda Sanitaria Universitaria Giuliano Isontina, 34125 Trieste, Italy; (G.B.S.); (C.F.)
| | - Carolina Fabiani
- Medicine of Services Department, Clinical Analysis Laboratory, Azienda Sanitaria Universitaria Giuliano Isontina, 34125 Trieste, Italy; (G.B.S.); (C.F.)
| | - Antonietta Robino
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (S.L.); (L.A.); (M.C.)
| | - Manola Comar
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (S.L.); (L.A.); (M.C.)
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy; (E.A.); (E.P.)
| |
Collapse
|
184
|
Mashhouri S, Rahmati A, Azimi A, Fava RA, Ismail IH, Walker J, Elahi S. Targeting Dectin-1 and or VISTA enhances anti-tumor immunity in melanoma but not colorectal cancer model. Cell Oncol (Dordr) 2024; 47:1735-1756. [PMID: 38668817 PMCID: PMC11467025 DOI: 10.1007/s13402-024-00950-w] [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] [Accepted: 04/11/2024] [Indexed: 06/27/2024] Open
Abstract
PURPOSE Acquired resistance to immune checkpoint blockers (ICBs) is a major barrier in cancer treatment, emphasizing the need for innovative strategies. Dectin-1 (gene Clec7a) is a C-type lectin receptor best known for its ability to recognize β-glucan-rich structures in fungal cell walls. While Dectin-1 is expressed in myeloid cells and tumor cells, its significance in cancer remains the subject of controversy. METHODS Using Celc7a-/- mice and curdlan administration to stimulate Dectin-1 signaling, we explored its impact. VISTA KO mice were employed to assess VISTA's role, and bulk RNAseq analyzed curdlan effects on neutrophils. RESULTS Our findings reveal myeloid cells as primary Dectin-1 expressing cells in the tumor microenvironment (TME), displaying an activated phenotype. Strong Dectin-1 co-expression/co-localization with VISTA and PD-L1 in TME myeloid cells was observed. While Dectin-1 deletion lacked protective effects, curdlan stimulation significantly curtailed B16-F10 tumor progression. RNAseq and pathway analyses supported curdlan's role in triggering a cascade of events leading to increased production of pro-inflammatory mediators, potentially resulting in the recruitment and activation of immune cells. Moreover, we identified a heterogeneous subset of Dectin-1+ effector T cells in the TME. Similar to mice, human myeloid cells are the prominent cells expressing Dectin-1 in cancer patients. CONCLUSION Our study proposes Dectin-1 as a potential adjunctive target with ICBs, orchestrating a comprehensive engagement of innate and adaptive immune responses in melanoma. This innovative approach holds promise for overcoming acquired resistance to ICBs in cancer treatment, offering avenues for further exploration and development.
Collapse
Affiliation(s)
- Siavash Mashhouri
- Department of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Amirhossein Rahmati
- Department of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Ako Azimi
- Department of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Roy A Fava
- Department of Veterans Affairs Medical Center, Research Service, White River Junction, VT, USA
- Department of Medicine, Geisel School of Medicine at Dartmouth, Dartmouth Hitchcock Medical Center, Lebanon, NH, USA
| | - Ismail Hassan Ismail
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- Biophysics Department, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - John Walker
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Shokrollah Elahi
- Department of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada.
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada.
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada.
| |
Collapse
|
185
|
Chen H, Huang S, Yao S, Wang J, Huang J, Yu Z. Multi-omics analyses of Bacillus amyloliquefaciens treated mice infected with Schistosoma japonicum reveal dynamics change of intestinal microbiome and its associations with host metabolism. PLoS Negl Trop Dis 2024; 18:e0012583. [PMID: 39466852 PMCID: PMC11515987 DOI: 10.1371/journal.pntd.0012583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 09/27/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Schistosomiasis japonica is a serious threat to human health. It causes damage to the intestine and liver. Probiotic therapy has been shown to be effective in alleviating intestinal diseases and improving host health. Previous studies have found that Bacillus amyloliquefaciens could alleviate the pathological symptoms of schistosomiasis japonica, but the regulatory mechanism of alleviating schistosomiasis japonica is still unknown. PRINCIPAL FINDINGS This study analyzed the dynamic changes of intestinal microbiome in mice infected with Schistosoma japonicum after the intervention of B. amyloliquefaciens and its connection to host metabolism by multi-omics sequencing technology. B. amyloliquefaciens was found to significantly regulate the homeostasis of intestinal microbiota by promoting the growth of beneficial bacteria and inhibiting potential pathogenic bacteria and protect the number of core microbes. Meanwhile, the genes related to the metabolism of glycerophospholipids and amino acid from intestinal microbiome changed significantly, and were shown to be significantly positively correlated with the associated metabolites of microbial origin. Moreover, host metabolism (lipid metabolism and steroid hormone biosynthesis) was also found to be significantly regulated. CONCLUSIONS The recovery of intestinal microbial homeostasis and the regulation of host metabolism revealed the potential probiotic properties of B. amyloliquefaciens, which also provided new ideas for the prevention and adjuvant treatment of schistosomiasis japonica.
Collapse
Affiliation(s)
- Hao Chen
- Human Microbiome and Health Group, Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Shuaiqin Huang
- Human Microbiome and Health Group, Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Siqi Yao
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Jingyan Wang
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Jing Huang
- Human Microbiome and Health Group, Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Zheng Yu
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| |
Collapse
|
186
|
Kang JW, Vemuganti V, Kuehn JF, Ulland TK, Rey FE, Bendlin BB. Gut microbial metabolism in Alzheimer's disease and related dementias. Neurotherapeutics 2024; 21:e00470. [PMID: 39462700 PMCID: PMC11585892 DOI: 10.1016/j.neurot.2024.e00470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/30/2024] [Accepted: 10/04/2024] [Indexed: 10/29/2024] Open
Abstract
Multiple studies over the last decade have established that Alzheimer's disease and related dementias (ADRD) are associated with changes in the gut microbiome. These alterations in organismal composition result in changes in the abundances of functions encoded by the microbial community, including metabolic capabilities, which likely impact host disease mechanisms. Gut microbes access dietary components and other molecules made by the host and produce metabolites that can enter circulation and cross the blood-brain barrier (BBB). In recent years, several microbial metabolites have been associated with or have been shown to influence host pathways relevant to ADRD pathology. These include short chain fatty acids, secondary bile acids, tryptophan derivatives (such as kynurenine, serotonin, tryptamine, and indoles), and trimethylamine/trimethylamine N-oxide. Notably, some of these metabolites cross the BBB and can have various effects on the brain, including modulating the release of neurotransmitters and neuronal function, inducing oxidative stress and inflammation, and impacting synaptic function. Microbial metabolites can also impact the central nervous system through immune, enteroendocrine, and enteric nervous system pathways, these perturbations in turn impact the gut barrier function and peripheral immune responses, as well as the BBB integrity, neuronal homeostasis and neurogenesis, and glial cell maturation and activation. This review examines the evidence supporting the notion that ADRD is influenced by gut microbiota and its metabolites. The potential therapeutic advantages of microbial metabolites for preventing and treating ADRD are also discussed, highlighting their potential role in developing new treatments.
Collapse
Affiliation(s)
- Jea Woo Kang
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Vaibhav Vemuganti
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Jessamine F Kuehn
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Tyler K Ulland
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Federico E Rey
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Barbara B Bendlin
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
187
|
Ratsika A, Codagnone MG, Bastiaanssen TFS, Hoffmann Sarda FA, Lynch CMK, Ventura-Silva AP, Rosell-Cardona C, Caputi V, Stanton C, Fülling C, Clarke G, Cryan JF. Maternal high-fat diet-induced microbiota changes are associated with alterations in embryonic brain metabolites and adolescent behaviour. Brain Behav Immun 2024; 121:317-330. [PMID: 39032541 DOI: 10.1016/j.bbi.2024.07.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 07/09/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024] Open
Abstract
The developing central nervous system is highly sensitive to nutrient changes during the perinatal period, emphasising the potential impact of alterations of maternal diet on offspring brain development and behaviour. A growing body of research implicates the gut microbiota in neurodevelopment and behaviour. Maternal overweight and obesity during the perinatal period has been linked to changes in neurodevelopment, plasticity and affective disorders in the offspring, with implications for microbial signals from the maternal gut. Here we investigate the impact of maternal high-fat diet (mHFD)-induced changes in microbial signals on offspring brain development, and neuroimmune signals, and the enduring effects on behaviour into adolescence. We first demonstrate that maternal caecal microbiota composition at term pregnancy (embryonic day 18: E18) differs significantly in response to maternal diet. Moreover, mHFD resulted in the upregulation of microbial genes in the maternal intestinal tissue linked to alterations in quinolinic acid synthesis and elevated kynurenine levels in the maternal plasma, both neuronal plasticity mediators related to glutamate metabolism. Metabolomics of mHFD embryonic brains at E18 also detected molecules linked to glutamate-glutamine cycle, including glutamic acid, glutathione disulphide, and kynurenine. During adolescence, the mHFD offspring exhibited increased locomotor activity and anxiety-like behaviour in a sex-dependent manner, along with upregulation of glutamate-related genes compared to controls. Overall, our results demonstrate that maternal exposure to high-fat diet results in microbiota changes, behavioural imprinting, altered brain metabolism, and glutamate signalling during critical developmental windows during the perinatal period.
Collapse
Affiliation(s)
- Anna Ratsika
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland
| | - Martin G Codagnone
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland
| | - Thomaz F S Bastiaanssen
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland
| | - Fabiana A Hoffmann Sarda
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Caoimhe M K Lynch
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland
| | - Ana Paula Ventura-Silva
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland
| | - Cristina Rosell-Cardona
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland
| | - Valentina Caputi
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland
| | | | - Christine Fülling
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork T12YT20, Ireland
| | - John F Cryan
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland.
| |
Collapse
|
188
|
Merali N, Chouari T, Sweeney C, Halle-Smith J, Jessel MD, Wang B, O’ Brien J, Suyama S, Jiménez JI, Roberts KJ, Velliou E, Sivakumar S, Rockall TA, Demirkan A, Pedicord V, Deng D, Giovannetti E, Annels NE, Frampton AE. The microbial composition of pancreatic ductal adenocarcinoma: a systematic review of 16S rRNA gene sequencing. Int J Surg 2024; 110:6771-6799. [PMID: 38874485 PMCID: PMC11487005 DOI: 10.1097/js9.0000000000001762] [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: 01/30/2024] [Accepted: 05/24/2024] [Indexed: 06/15/2024]
Abstract
BACKGROUND Pancreatic cancer, specifically pancreatic ductal adenocarcinoma (PDAC), continues to pose a significant clinical and scientific challenge. The most significant finding of recent years is that PDAC tumours harbour their specific microbiome, which differs amongst tumour entities and is distinct from healthy tissue. This review aims to evaluate and summarise all PDAC studies that have used the next-generation technique, 16S rRNA gene amplicon sequencing within each bodily compartment. As well as establishing a causal relationship between PDAC and the microbiome. MATERIALS AND METHODS This systematic review was carried out according to the Preferred Reporting Items for Systematic Reviews and Meta-analysis (PRISMA) guidelines. A comprehensive search strategy was designed, and 1727 studies were analysed. RESULTS In total, 38 studies were selected for qualitative analysis and summarised significant PDAC bacterial signatures. Despite the growing amount of data provided, we are not able to state a universal 16S rRNA gene microbial signature that can be used for PDAC screening. This is most certainly due to the heterogeneity of the presentation of results, lack of available datasets, and the intrinsic selection bias between studies. CONCLUSION Several key studies have begun to shed light on causality and the influence the microbiome constituents and their produced metabolites could play in tumorigenesis and influencing outcomes. The challenge in this field is to shape the available microbial data into targetable signatures. Making sequenced data readily available is critical, coupled with the coordinated standardisation of data and the need for consensus guidelines in studies investigating the microbiome in PDAC.
Collapse
Affiliation(s)
- Nabeel Merali
- Minimal Access Therapy Training Unit (MATTU), Royal Surrey Hospital NHS Foundation Trust
- Department of Hepato-Pancreato-Biliary (HPB) Surgery, Royal Surrey County Hospital NHS Foundation Trust
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey
| | - Tarak Chouari
- Minimal Access Therapy Training Unit (MATTU), Royal Surrey Hospital NHS Foundation Trust
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey
| | - Casie Sweeney
- Minimal Access Therapy Training Unit (MATTU), Royal Surrey Hospital NHS Foundation Trust
| | - James Halle-Smith
- Hepatobiliary and Pancreatic Surgery Unit, Queen Elizabeth Hospital Birmingham, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Maria-Danae Jessel
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey
| | - Bing Wang
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam
| | - James O’ Brien
- Minimal Access Therapy Training Unit (MATTU), Royal Surrey Hospital NHS Foundation Trust
| | - Satoshi Suyama
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge
| | | | - Keith J. Roberts
- Hepatobiliary and Pancreatic Surgery Unit, Queen Elizabeth Hospital Birmingham, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Eirini Velliou
- Centre for 3D Models of Health and Disease, Division of Surgery and Interventional Science, University College London (UCL), London
| | - Shivan Sivakumar
- Oncology Department and Institute of Immunology and Immunotherapy, Birmingham Medical School, University of Birmingham
| | - Timothy A. Rockall
- Minimal Access Therapy Training Unit (MATTU), Royal Surrey Hospital NHS Foundation Trust
| | - Ayse Demirkan
- Section of Statistical Multi-Omics, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey
- Surrey Institute for People-Centred AI, University of Surrey, Guildford, Surrey
| | - Virginia Pedicord
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge
| | - Dongmei Deng
- Department of Preventive Dentistry, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Elisa Giovannetti
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam
- Fondazione Pisa per la Scienza, San Giuliano, Italy
| | - Nicola E. Annels
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey
| | - Adam E. Frampton
- Minimal Access Therapy Training Unit (MATTU), Royal Surrey Hospital NHS Foundation Trust
- Department of Hepato-Pancreato-Biliary (HPB) Surgery, Royal Surrey County Hospital NHS Foundation Trust
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey
| |
Collapse
|
189
|
Neufeld PM, Nettersheim RA, Matschke V, Vorgerd M, Stahlke S, Theiss C. Unraveling the gut-brain axis: the impact of steroid hormones and nutrition on Parkinson's disease. Neural Regen Res 2024; 19:2219-2228. [PMID: 38488556 PMCID: PMC11034592 DOI: 10.4103/1673-5374.391304] [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: 05/23/2023] [Revised: 10/07/2023] [Accepted: 11/24/2023] [Indexed: 04/24/2024] Open
Abstract
This comprehensive review explores the intricate relationship between nutrition, the gut microbiome, steroid hormones, and Parkinson's disease within the context of the gut-brain axis. The gut-brain axis plays a pivotal role in neurodegenerative diseases like Parkinson's disease, encompassing diverse components such as the gut microbiota, immune system, metabolism, and neural pathways. The gut microbiome, profoundly influenced by dietary factors, emerges as a key player. Nutrition during the first 1000 days of life shapes the gut microbiota composition, influencing immune responses and impacting both child development and adult health. High-fat, high-sugar diets can disrupt this delicate balance, contributing to inflammation and immune dysfunction. Exploring nutritional strategies, the Mediterranean diet's anti-inflammatory and antioxidant properties show promise in reducing Parkinson's disease risk. Microbiome-targeted dietary approaches and the ketogenic diet hold the potential in improving brain disorders. Beyond nutrition, emerging research uncovers potential interactions between steroid hormones, nutrition, and Parkinson's disease. Progesterone, with its anti-inflammatory properties and presence in the nervous system, offers a novel option for Parkinson's disease therapy. Its ability to enhance neuroprotection within the enteric nervous system presents exciting prospects. The review addresses the hypothesis that α-synuclein aggregates originate from the gut and may enter the brain via the vagus nerve. Gastrointestinal symptoms preceding motor symptoms support this hypothesis. Dysfunctional gut-brain signaling during gut dysbiosis contributes to inflammation and neurotransmitter imbalances, emphasizing the potential of microbiota-based interventions. In summary, this review uncovers the complex web of interactions between nutrition, the gut microbiome, steroid hormones, and Parkinson's disease within the gut-brain axis framework. Understanding these connections not only offers novel therapeutic insights but also illuminates the origins of neurodegenerative diseases such as Parkinson's disease.
Collapse
Affiliation(s)
- Paula Maria Neufeld
- Department of Cytology, Institute of Anatomy, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
| | - Ralf A. Nettersheim
- Department of Visceral Surgery, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany
| | - Veronika Matschke
- Department of Cytology, Institute of Anatomy, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
| | - Matthias Vorgerd
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany
| | - Sarah Stahlke
- Department of Cytology, Institute of Anatomy, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
| | - Carsten Theiss
- Department of Cytology, Institute of Anatomy, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
190
|
Moles L, Otaegui-Chivite A, Gorostidi-Aicua M, Romarate L, Mendiburu I, Crespillo-Velasco H, Álvarez de Arcaya A, Ferreira E, Arruti M, Castillo-Triviño T, Otaegui D. Microbiota modulation by teriflunomide therapy in people with multiple sclerosis: An observational case-control study. Neurotherapeutics 2024; 21:e00457. [PMID: 39406600 PMCID: PMC11585876 DOI: 10.1016/j.neurot.2024.e00457] [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: 07/01/2024] [Revised: 09/19/2024] [Accepted: 09/21/2024] [Indexed: 11/16/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic immune-mediated and heterogeneous disease characterized by demyelination, axonal damage, and physical and cognitive impairment. Recent studies have highlighted alterations in the microbiota of people with MS (pwMS). However, the intricate nature of the disease and the wide range of treatments available make it challenging to identify specific microbial populations or functions associated with MS symptoms and disease progression. This study aimed to characterize the microbiota of pwMS treated with the oral drug teriflunomide (TF) and compare it with that of pwMS treated with beta interferons (IFNβ), pwMS treated with no previous disease modifying therapies (naïve), and healthy controls. Our findings demonstrate significant alterations in both the composition and function of the gut microbiota in pwMS that are further influenced by disease-modifying therapies. Specifically, oral treatment with TF had a notable impact on the gut microbiota of pwMS. Importantly, the dysregulated microbial environment within the gut was associated with symptoms commonly experienced by pwMS, including fatigue, anxiety, and depression.
Collapse
Affiliation(s)
- Laura Moles
- Biogipuzkoa Health Research Institute, Group of Neuroimmunilogy, San Sebastián, Spain; Center for Biomedical Research Network in Neurodegenerative Diseases (CIBER-CIBERNED-ISCIII), Madrid, Spain
| | - Ane Otaegui-Chivite
- Biogipuzkoa Health Research Institute, Group of Neuroimmunilogy, San Sebastián, Spain; Center for Biomedical Research Network in Neurodegenerative Diseases (CIBER-CIBERNED-ISCIII), Madrid, Spain
| | - Miriam Gorostidi-Aicua
- Biogipuzkoa Health Research Institute, Group of Neuroimmunilogy, San Sebastián, Spain; Center for Biomedical Research Network in Neurodegenerative Diseases (CIBER-CIBERNED-ISCIII), Madrid, Spain
| | - Leire Romarate
- Biogipuzkoa Health Research Institute, Group of Neuroimmunilogy, San Sebastián, Spain
| | - Idoia Mendiburu
- Biogipuzkoa Health Research Institute, Group of Neuroimmunilogy, San Sebastián, Spain; Neurology Department, Hospital Universitario Donostia, Osakidetza Basque Health Service, San Sebastián, Spain
| | | | - Amaya Álvarez de Arcaya
- Neurology Department, Hospital Universitario Araba, Osakidetza Basque Health Service, Vitoria-Gasteiz, Spain
| | - Eva Ferreira
- Neurology Department, Hospital Universitario Araba, Osakidetza Basque Health Service, Vitoria-Gasteiz, Spain
| | - Maialen Arruti
- Biogipuzkoa Health Research Institute, Group of Neuroimmunilogy, San Sebastián, Spain; Neurology Department, Hospital Universitario Donostia, Osakidetza Basque Health Service, San Sebastián, Spain
| | - Tamara Castillo-Triviño
- Biogipuzkoa Health Research Institute, Group of Neuroimmunilogy, San Sebastián, Spain; Center for Biomedical Research Network in Neurodegenerative Diseases (CIBER-CIBERNED-ISCIII), Madrid, Spain; Neurology Department, Hospital Universitario Donostia, Osakidetza Basque Health Service, San Sebastián, Spain
| | - David Otaegui
- Biogipuzkoa Health Research Institute, Group of Neuroimmunilogy, San Sebastián, Spain; Center for Biomedical Research Network in Neurodegenerative Diseases (CIBER-CIBERNED-ISCIII), Madrid, Spain.
| |
Collapse
|
191
|
Zhang SL, Wang X, Cai QQ, Chen C, Zhang ZY, Xu YY, Yang MX, Jia QA, Wang Y, Wang ZM. Acarbose enhances the efficacy of immunotherapy against solid tumours by modulating the gut microbiota. Nat Metab 2024; 6:1991-2009. [PMID: 39322747 DOI: 10.1038/s42255-024-01137-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 08/29/2024] [Indexed: 09/27/2024]
Abstract
The crucial role of gut microbiota in shaping immunotherapy outcomes has prompted investigations into potential modulators. Here we show that oral administration of acarbose significantly increases the anti-tumour response to anti-PD-1 therapy in female tumour-bearing mice. Acarbose modulates the gut microbiota composition and tryptophan metabolism, thereby contributing to changes in chemokine expression and increased T cell infiltration within tumours. We identify CD8+ T cells as pivotal components determining the efficacy of the combined therapy. Further experiments reveal that acarbose promotes CD8+ T cell recruitment through the CXCL10-CXCR3 pathway. Faecal microbiota transplantation and gut microbiota depletion assays indicate that the effects of acarbose are dependent on the gut microbiota. Specifically, acarbose enhances the efficacy of anti-PD-1 therapy via the tryptophan catabolite indoleacetate, which promotes CXCL10 expression and thus facilitates CD8+ T cell recruitment, sensitizing tumours to anti-PD-1 therapy. The bacterial species Bifidobacterium infantis, which is enriched by acarbose, also improves response to anti-PD-1 therapy. Together, our study endorses the potential combination of acarbose and anti-PD-1 for cancer immunotherapy.
Collapse
Affiliation(s)
- Shi-Long Zhang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, P. R. China.
| | - Xin Wang
- Department of Integrative Medicine, Shanghai Geriatric Center, Minhang District, Shanghai, P.R. China
| | - Qing-Qing Cai
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Chen Chen
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Zheng-Yan Zhang
- State Key Laboratory of Oncogenes and Related Genes, Stem Cell Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| | - Ya-Yun Xu
- Department of Biliary-Pancreatic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| | - Meng-Xuan Yang
- Department of Gastrointestinal Surgery, Minhang hospital, Fudan University, Shanghai, P. R. China
| | - Qing-An Jia
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, P. R. China.
| | - Yan Wang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, P. R. China.
| | - Zhi-Ming Wang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, P. R. China.
- Department of Medical Oncology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, P. R. China.
| |
Collapse
|
192
|
Dandamudi BJ, Dimaano KAM, Shah N, AlQassab O, Al-Sulaitti Z, Nelakuditi B, Mohammed L. Neurodegenerative Disorders and the Gut-Microbiome-Brain Axis: A Literature Review. Cureus 2024; 16:e72427. [PMID: 39588438 PMCID: PMC11588320 DOI: 10.7759/cureus.72427] [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: 09/03/2024] [Accepted: 10/26/2024] [Indexed: 11/27/2024] Open
Abstract
Neurodegenerative diseases are severe, age-related conditions with complex etiologies that result in significant morbidity and mortality. The gut microbiome, a dynamic symbiotic environment comprising commensal organisms, represents the largest reservoir of these organisms within the human body. It produces short-chain fatty acids, endogenous signals, and neuroactive compounds, which can modulate neuronal function, plasticity, and behavior. Emerging evidence suggests that the gut microbiome plays a pivotal role in neurodevelopment, aging, and brain diseases, including Alzheimer's disease, Parkinson's disease, and stroke. Communication between the gut and brain occurs through a bidirectional channel known as the gut-microbiome-brain axis, which is being explored for therapeutic potential in neurodegenerative disorders. This literature review was conducted through a comprehensive search of five electronic databases - PubMed, Scopus, Ovid Medline, Cochrane Review, and Google Scholar - from inception to June 2024, focusing on English-language studies. Keywords included "gut-brain axis", "microbiome dysbiosis", "neurodegeneration", and disorder-specific terms such as "Alzheimer's disease" and "Parkinson's disease", paired with "gut microbiome". The review examines current knowledge on the relationship between gut microbiota and neurodegenerative disorders, emphasizing potential mechanisms and therapeutic options. Results indicate that gut dysbiosis, characterized by microbial imbalance, is intricately associated with neurodegenerative disease pathogenesis by influencing immune responses, increasing blood-brain barrier permeability, and generating neurotoxic metabolites. Therapeutic approaches targeting the gut microbiome, including probiotics, prebiotics, and fecal microbiota transplantation, show promise in restoring microbial balance and slowing disease progression. However, further research is essential to validate these findings and develop effective clinical interventions.
Collapse
Affiliation(s)
- Bindu Jyothi Dandamudi
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Kathrina Antheia M Dimaano
- Obstetrics and Gynecology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Nensi Shah
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Osamah AlQassab
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Zainab Al-Sulaitti
- Obstetrics and Gynecology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Bhavana Nelakuditi
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Lubna Mohammed
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
193
|
Mahmoudi F, Jalayeri MHT, Montaseri A, MohamedKhosroshahi L, Baradaran B. Microbial natural compounds and secondary metabolites as Immunomodulators: A review. Int J Biol Macromol 2024; 278:134778. [PMID: 39153680 DOI: 10.1016/j.ijbiomac.2024.134778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 08/11/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Immunomodulatory therapies are beneficial strategies for the improvement of immune system function. Today, due to the increasing prevalence of immune disorders, cancer, and new viral diseases, there is a greater need to introduce immunomodulatory compounds with more efficiency and fewer side effects. Microbial derivatives are fertile and attractive grounds for discovering lots of novel compounds with various medical properties. The discovery of many natural compounds derived from bacterial sources, such as secondary metabolites with promising immunomodulating activities, represents the importance of this topic in drug discovery and emphasizes the necessity for a coherent source of study in this area. Considering this need, in this review, we aim to focus on the current information about the immunomodulatory effects of bacterial secondary metabolites and natural immunomodulators derived from microorganisms.
Collapse
Affiliation(s)
- Fariba Mahmoudi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad Hadi Tajik Jalayeri
- Clinical Research Development Unit (CRDU), Sayad Shirazi Hospital Golestan University of Medical Sciences, Gorgan, Iran
| | - Azadeh Montaseri
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy.
| | - Leila MohamedKhosroshahi
- Department of Immunology, School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
194
|
Kawakami N, Wekerle H. Life history of a brain autoreactive T cell: From thymus through intestine to blood-brain barrier and brain lesion. Neurotherapeutics 2024; 21:e00442. [PMID: 39237437 PMCID: PMC11585894 DOI: 10.1016/j.neurot.2024.e00442] [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: 05/06/2024] [Revised: 08/21/2024] [Accepted: 08/21/2024] [Indexed: 09/07/2024] Open
Abstract
Brain antigen-specific autoreactive T cells seem to play a key role in inducing inflammation in the central nervous system (CNS), a characteristic feature of human multiple sclerosis (MS). These T cells are generated within the thymus, where they escape negative selection and become integrated into the peripheral immune repertoire of immune cells. Typically, these autoreactive T cells rest in the periphery without attacking the CNS. When autoimmune T cells enter gut-associated lymphatic tissue (GALT), they may be stimulated by the microbiota and its metabolites. After activation, the cells migrate into the CNS through the blood‒brain barrier, become reactivated upon interacting with local antigen-presenting cells, and induce inflammatory lesions within the brain parenchyma. This review describes how microbiota influence autoreactive T cells during their life, starting in the thymus, migrating through the periphery and inducing inflammation in their target organ, the CNS.
Collapse
Affiliation(s)
- Naoto Kawakami
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich and Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Germany.
| | - Hartmut Wekerle
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich and Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Germany; Emeritus Group Neuroimmunology, Max Planck Institute of Biological Intelligence, Germany.
| |
Collapse
|
195
|
Qusty N, Sarhan A, Taha M, Alshanqiti A, Almuteb AM, Alfaraidi AT, Alkhairi HA, Alzahrani MM, Alamry AHA, Alomry TQB, Bannan OA, Almaashi MS. The Role of Gut Microbiota in the Efficacy and Side Effect Profile of Biologic Therapies for Autoimmune Diseases. Cureus 2024; 16:e71111. [PMID: 39525264 PMCID: PMC11548951 DOI: 10.7759/cureus.71111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2024] [Indexed: 11/16/2024] Open
Abstract
The role of gut microbiota in influencing the efficacy and side effect profile of biological therapies for autoimmune diseases has gained increasing attention. Understanding these interactions is crucial for optimizing treatment outcomes and minimizing adverse events associated with biological therapies. This systematic review was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. We comprehensively analyzed studies involving human subjects with autoimmune diseases treated with biological therapies. Data on gut microbiota composition, therapeutic efficacy, and side effect profiles were extracted and synthesized to assess the impact of microbiota on treatment outcomes. Our review identified a significant relationship between gut microbiota composition and the efficacy of biological therapies. Specific bacterial taxa, such as Clostridiales and Roseburia inulinivorans, were associated with improved therapeutic responses, while alterations in microbiota were linked to increased adverse events. The predictive potential was demonstrated with microbiota signatures correlating with treatment success and side effects, highlighting the relevance of microbial profiles in therapeutic outcomes. The findings suggest that gut microbiota plays a pivotal role in modulating the efficacy and side effect profile of biological therapies for autoimmune diseases. Integrating microbiota assessments into clinical practice could enhance personalized treatment strategies and improve patient outcomes.
Collapse
Affiliation(s)
- Naeem Qusty
- Department of Clinical Laboratory Sciences, Umm Al-Qura University, Makkah, SAU
| | - Anas Sarhan
- Department of Internal Medicine, Umm Al-Qura University, Makkah, SAU
| | - Medhat Taha
- Department of Anatomy, Umm Al-Qura University, Al-Qunfudhah, SAU
| | | | | | | | | | | | | | | | | | | |
Collapse
|
196
|
Nichols RG, Rimal B, Hao F, Peters JM, Davenport ER, Patterson AD. Chlorpyrifos modulates the mouse gut microbiota and metabolic activity. ENVIRONMENT INTERNATIONAL 2024; 192:109022. [PMID: 39312839 PMCID: PMC11583244 DOI: 10.1016/j.envint.2024.109022] [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: 02/19/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 09/25/2024]
Abstract
The organophosphate chlorpyrifos is a commonly used pesticide for fruits and vegetables despite its association with neurotoxicity in humans. While some studies suggest that organophosphates may impact the gut microbiota, no studies to date have investigated the direct effect of chlorpyrifos on the gut microbiota with doses that approximate environmentally relevant dietary concentrations (EPA chronic reference dose: 0.3 µg/kg/day in humans and EPA acute reference dose: 5 µg/kg/day in humans). Thus, we examined the influence of chlorpyrifos on the gut microbiota by assessment of bacterial physiology and metabolism using flow cytometry, 1H NMR-based metabolomics, and changes in the cecal microbiota community with 16S rRNA amplicon sequencing and analysis. Chlorpyrifos did not directly damage bacteria but rather perturbed bacterial metabolism. Chlorpyrifos exposure to bacteria increased the concentration of amino acids, carbohydrates, and nucleic acids. The relative abundances of Lactobacillus, Allobaculum, Roseburia, and Butyricicoccus increased after exposure to chlorpyrifos. Analyses of the 16S rRNA gene amplicon data predicted decreased amino acid biosynthesis and nucleic acid degradation and increased glycolysis which was supported by 1H NMR-based metabolomics. Collectively, these results demonstrate that environmentally relevant doses of chlorpyrifos can impact the metabolic activity of isolated gut microbes which may result in an imbalance in overall gut metabolic activity.
Collapse
Affiliation(s)
- Robert G Nichols
- Department of Biology, The Pennsylvania State University, University Park, PA, USA; One Health Microbiome Center, Huck Life Sciences Institute, University Park, PA 16802, USA
| | - Bipin Rimal
- One Health Microbiome Center, Huck Life Sciences Institute, University Park, PA 16802, USA; Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA, USA
| | - Fuhua Hao
- Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA, USA
| | - Jeffrey M Peters
- Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA, USA
| | - Emily R Davenport
- Department of Biology, The Pennsylvania State University, University Park, PA, USA; One Health Microbiome Center, Huck Life Sciences Institute, University Park, PA 16802, USA
| | - Andrew D Patterson
- One Health Microbiome Center, Huck Life Sciences Institute, University Park, PA 16802, USA; Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
197
|
Leclaire S, Bandekar M, Rowe M, Ritari J, Jokiniemi A, Partanen J, Allinen P, Kuusipalo L, Kekäläinen J. Female reproductive tract microbiota varies with MHC profile. Proc Biol Sci 2024; 291:20241334. [PMID: 39471862 PMCID: PMC11521592 DOI: 10.1098/rspb.2024.1334] [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: 06/03/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 11/01/2024] Open
Abstract
Numerous studies have shown that a healthy reproductive tract microbiota is crucial for successful reproduction and that its composition is influenced by various environmental and host factors. However, it is not known whether the reproductive microbiota is also shaped by the major histocompatibility complex (MHC), a family of genes essential to differentiate 'self' from 'non-self' peptides to initiate an adaptive immune response. We tested the association between the follicular fluid microbiome and MHC genes in 27 women. Women with higher MHC diversity had a higher microbiome diversity, characterized by bacteria commonly associated with vaginal dysbiosis. Women with similar MHC genes were also similar in their microbiome composition, indicating that MHC composition may be a key factor in determining the bacterial assemblage in the reproductive tract. Finally, the composition of the follicular fluid microbiome was similar to the vaginal microbiome, suggesting that numerous bacteria of the vagina are true inhabitants of the follicular fluid or that vaginal microbiota contaminated the follicular fluid microbiota during transvaginal collection. Collectively, our results demonstrate the importance of host genetic factors in shaping women's reproductive microbiota and they open the door for further research on the role of microbiota in mediating MHC-related variation in reproductive success.
Collapse
Affiliation(s)
- Sarah Leclaire
- Centre de Recherche sur la Biodiversité et l’Environnement (CRBE), UMR5300, Université Toulouse, CNRS, IRD, Toulouse INP, 118 rte de Narbonne, Toulouse31062, France
| | - Mandar Bandekar
- Department of Environmental and Biological Sciences, University of Eastern Finland, PO Box 111, Joensuu80101, Finland
| | - Melissah Rowe
- Department of Animal Ecology, Netherlands Institute of Ecology (NIOO-KNAW), Wageningen6700 AB, The Netherlands
| | - Jarmo Ritari
- Finnish Red Cross Blood Service, Research and Development, Haartmaninkatu 8, Helsinki00290, Finland
| | - Annalaura Jokiniemi
- Department of Environmental and Biological Sciences, University of Eastern Finland, PO Box 111, Joensuu80101, Finland
| | - Jukka Partanen
- Finnish Red Cross Blood Service, Research and Development, Haartmaninkatu 8, Helsinki00290, Finland
| | - Pia Allinen
- Ovumia Kuopio, Ajurinkatu 16, Kuopio70110, Finland
| | - Liisa Kuusipalo
- North Karelia Central Hospital, Tikkamäentie 16, Joensuu80210, Finland
| | - Jukka Kekäläinen
- Department of Environmental and Biological Sciences, University of Eastern Finland, PO Box 111, Joensuu80101, Finland
| |
Collapse
|
198
|
Yadav S, Sapra L, Srivastava RK. Polysaccharides to postbiotics: Nurturing bone health via modulating "gut-immune axis". Int J Biol Macromol 2024; 278:134655. [PMID: 39128750 DOI: 10.1016/j.ijbiomac.2024.134655] [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: 01/11/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
The increasing prevalence of individuals affected by bone pathologies globally has sparked catastrophic concerns. Ankylosing spondylitis, osteoporosis, rheumatoid arthritis, osteoarthritis, and fractures alone impact an estimated 1.71 billion people worldwide. The gut microbiota plays a crucial role in interacting with the host through the synthesis of a diverse range of metabolites called gut-associated metabolites (GAMs), which originate from external dietary substrates or endogenous host compounds. Many metabolic disorders have been linked to alterations in the gut microbiota's activity and composition. The development of metabolic illnesses has been linked to certain microbiota-derived metabolites, such as branched-chain amino acids, bile acids, short-chain fatty acids, tryptophan, trimethylamine N-oxide, and indole derivatives. Moreover, the modulation of gut microbiota through biotics (prebiotics, probiotics and postbiotics) presents a promising avenue for therapeutic intervention. Biotics selectively promote the growth of beneficial gut bacteria, thereby enhancing the production of GAMs with potential beneficial effects on bone metabolism. Understanding the intricate interplay between GAMs, and bone-associated genes through molecular informatics holds significant promise for early diagnosis, prognosis, and novel treatment strategies for various bone disorders.
Collapse
Affiliation(s)
- Sumedha Yadav
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Leena Sapra
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Rupesh K Srivastava
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India.
| |
Collapse
|
199
|
Schneider KM, Kummen M, Trivedi PJ, Hov JR. Role of microbiome in autoimmune liver diseases. Hepatology 2024; 80:965-987. [PMID: 37369002 PMCID: PMC11407779 DOI: 10.1097/hep.0000000000000506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/25/2023] [Indexed: 06/29/2023]
Abstract
The microbiome plays a crucial role in integrating environmental influences into host physiology, potentially linking it to autoimmune liver diseases, such as autoimmune hepatitis, primary biliary cholangitis, and primary sclerosing cholangitis. All autoimmune liver diseases are associated with reduced diversity of the gut microbiome and altered abundance of certain bacteria. However, the relationship between the microbiome and liver diseases is bidirectional and varies over the course of the disease. This makes it challenging to dissect whether such changes in the microbiome are initiating or driving factors in autoimmune liver diseases, secondary consequences of disease and/or pharmacological intervention, or alterations that modify the clinical course that patients experience. Potential mechanisms include the presence of pathobionts, disease-modifying microbial metabolites, and more nonspecific reduced gut barrier function, and it is highly likely that the effect of these change during the progression of the disease. Recurrent disease after liver transplantation is a major clinical challenge and a common denominator in these conditions, which could also represent a window to disease mechanisms of the gut-liver axis. Herein, we propose future research priorities, which should involve clinical trials, extensive molecular phenotyping at high resolution, and experimental studies in model systems. Overall, autoimmune liver diseases are characterized by an altered microbiome, and interventions targeting these changes hold promise for improving clinical care based on the emerging field of microbiota medicine.
Collapse
Affiliation(s)
| | - Martin Kummen
- Norwegian PSC Research Center, Department of Transplantation Medicine, Oslo University Hospital Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Palak J. Trivedi
- National Institute for Health and Care Research Birmingham Biomedical Research Centre, Centre for Liver and Gastroenterology Research, University of Birmingham, UK
- Liver Unit, University Hospitals Birmingham Queen Elizabeth, Birmingham, UK
- Institute of Immunology and Immunotherapy, University of Birmingham, UK
- Institute of Applied Health Research, University of Birmingham, UK
| | - Johannes R. Hov
- Norwegian PSC Research Center, Department of Transplantation Medicine, Oslo University Hospital Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Section of Gastroenterology, Department of Transplantation Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| |
Collapse
|
200
|
Lei ZN, Teng QX, Koya J, Liu Y, Chen Z, Zeng L, Chen ZS, Fang S, Wang J, Liu Y, Pan Y. The correlation between cancer stem cells and epithelial-mesenchymal transition: molecular mechanisms and significance in cancer theragnosis. Front Immunol 2024; 15:1417201. [PMID: 39403386 PMCID: PMC11471544 DOI: 10.3389/fimmu.2024.1417201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 09/06/2024] [Indexed: 01/03/2025] Open
Abstract
The connections between cancer stem cells (CSCs) and epithelial-mesenchymal transition (EMT) is critical in cancer initiation, progression, metastasis, and therapy resistance, making it a focal point in cancer theragnosis. This review provides a panorama of associations and regulation pathways between CSCs and EMT, highlighting their significance in cancer. The molecular mechanisms underlined EMT are thoroughly explored, including the involvement of key transcription factors and signaling pathways. In addition, the roles of CSCs and EMT in tumor biology and therapy resistance, is further examined in this review. The clinical implications of CSCs-EMT interplay are explored, including identifying mesenchymal-state CSC subpopulations using advanced research methods and developing targeted therapies such as inhibitors and combination treatments. Overall, understanding the reciprocal relationship between EMT and CSCs holds excellent potential for informing the development of personalized therapies and ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Zi-Ning Lei
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, New York, NY, United States
| | - Qiu-Xu Teng
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, New York, NY, United States
| | - Jagadish Koya
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, New York, NY, United States
| | - Yangruiyu Liu
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Zizhou Chen
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Leli Zeng
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, New York, NY, United States
| | - Shuo Fang
- Big Data Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
- Department of Oncology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Jinxiang Wang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yuchen Liu
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
- Big Data Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yihang Pan
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
- Big Data Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| |
Collapse
|