1
|
Thakur P, Baraskar K, Shrivastava VK, Medhi B. Cross-talk between adipose tissue and microbiota-gut-brain-axis in brain development and neurological disorder. Brain Res 2024; 1844:149176. [PMID: 39182900 DOI: 10.1016/j.brainres.2024.149176] [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: 04/12/2024] [Revised: 07/25/2024] [Accepted: 08/18/2024] [Indexed: 08/27/2024]
Abstract
The gut microbiota is an important factor responsible for the physiological processes as well as pathogenesis of host. The communication between central nervous system (CNS) and microbiota occurs by different pathways i.e., chemical, neural, immune, and endocrine. Alteration in gut microbiota i.e., gut dysbiosis causes alteration in the bidirectional communication between CNS and gut microbiota and linked to the pathogenesis of neurological and neurodevelopmental disorder. Therefore, now-a-days microbiota-gut-brain-axis (MGBA) has emerged as therapeutic target for the treatment of metabolic disorder. But, experimental data available on MGBA from basic research has limited application in clinical study. In present study we first summarized molecular mechanism of microbiota interaction with brain physiology and pathogenesis via collecting data from different sources i.e., PubMed, Scopus, Web of Science. Furthermore, evidence shows that adipose tissue (AT) is active during metabolic activities and may also interact with MGBA. Hence, in present study we have focused on the relationship among MGBA, brown adipose tissue, and white adipose tissue. Along with this, we have also studied functional specificity of AT, and understanding heterogeneity among MGBA and different types of AT. Therefore, molecular interaction among them may provide therapeutic target for the treatment of neurological disorder.
Collapse
Affiliation(s)
- Pratibha Thakur
- Endocrinology Unit, Bioscience Department, Barkatullah University, Bhopal, Madhya Pradesh 462026, India.
| | - Kirti Baraskar
- Endocrinology Unit, Bioscience Department, Barkatullah University, Bhopal, Madhya Pradesh 462026, India
| | - Vinoy K Shrivastava
- Endocrinology Unit, Bioscience Department, Barkatullah University, Bhopal, Madhya Pradesh 462026, India
| | - Bikash Medhi
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, Punjab 160012, India.
| |
Collapse
|
2
|
Guo YF, Liu ZY, Zhou M, Kuang WH, Liu Y, Huang Y, Yin P, Xia ZY. Heat exposure promotes sarcopenia via gut microbiota-derived metabolites. Aging Cell 2024:e14370. [PMID: 39468887 DOI: 10.1111/acel.14370] [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: 07/01/2024] [Revised: 09/18/2024] [Accepted: 09/24/2024] [Indexed: 10/30/2024] Open
Abstract
The unprecedented rise in global ambient temperatures in the last decade has significantly impacted human health, yet how heat exposure affects the development of sarcopenia remains enigmatic. Here, we demonstrate that chronic heat exposure induces skeletal muscle volume loss, leading to muscle strength and functional decline in mice. The microbiota composition of heat-exposed mice was analyzed using 16S ribosomal DNA analysis. Liquid chromatography-mass spectrometry (LC-MS) was used to explore the effects of heat exposure on the blood metabolome and to further analyze the correlation between blood metabolism and gut microbiota. Transplantation of microbiota from heat-exposed mice to germ-free mice was sufficient to increase adverse effects on skeletal muscle function in the host. Mechanistically, using an untargeted metabolomics strategy, we reveal that altered gut microbiota due to high temperatures is associated with elevated serum levels of homocitrulline. Homocitrulline causes mitochondrial dysfunction in myocytes by exacerbating ferroptosis levels. And Nrf2 activator (Oltipraz) supplementation alleviates muscle atrophy and dysfunction induced by heat exposure. Our findings reveal the detrimental effects of heat exposure on muscle function and provide new strategies for treating sarcopenia.
Collapse
Affiliation(s)
- Yi-Fan Guo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Zhe-Yu Liu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Min Zhou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Wei-Hong Kuang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Ya Liu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yan Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Ping Yin
- Department of Oral and Maxillofacial Surgery, Center of Stomatology,Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhu-Ying Xia
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
3
|
Guo J, Li Z, Liu X, Jin Y, Sun Y, Yuan Z, Zhang W, Wang J, Zhang M. Response of the gut microbiota to changes in the nutritional status of red deer during winter. Sci Rep 2024; 14:24961. [PMID: 39438539 PMCID: PMC11496518 DOI: 10.1038/s41598-024-76142-1] [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: 05/15/2024] [Accepted: 10/10/2024] [Indexed: 10/25/2024] Open
Abstract
Unravelling abrupt alterations in the gut microbiota of wild species associated with nutritional stress is imperative but challenging for wildlife conservation. This study assessed the nutritional status of wild red deer during winter on the basis of changes in faecal nitrogen (FN) and urea nitrogen/creatinine (UN: C) levels and identified gut microbes associated with nutritional status via nutritional control experiments and metagenomic sequencing. The FN of wild red deer in winter 2022 was significantly lower than that in winter 2021 (p < 0.05, winter 2021: 1.37 ± 0.16% and winter 2022: 1.26 ± 0.22%), and the UN: C ratio increased (winter 2021: 2.19 ± 1.65 and winter 2022: 3.05 ± 3.50). Similar trends were found in late winter, which indicated greater nutritional pressure in winter (2022) and late winter. Compared with winter 2021, abundances of Ructibacterium and Butyrivibrio significantly increased, and Acetatifactor and Cuneatibacter significantly decreased during winter 2022 (p < 0.05). Compared with early winter, the cell growth and death pathways increased and lipid metabolism and its subpathway of secondary bile acid synthesis (ko00121) significantly decreased during late winter (p < 0.05), which was similar to the changes in malnourished experimental red deer. Abrupt alterations in the gut microbiota should receive increased attention when monitoring the nutritional health of wild ungulates. This study provides new insights and critical implications for the conservation of wild ungulate populations.
Collapse
Affiliation(s)
- Jinhao Guo
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, 150040, China
| | - Zheng Li
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, 100083, China
| | - Xinxin Liu
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, 150040, China
| | - Yongchao Jin
- Forestry and Grassland College, Jilin Agricultural University, Changchun, 130118, China
- World Wild Fund for Nature, Beijing, 100009, China
| | - Yue Sun
- School of Biological Sciences, Guizhou Education University, Guiyang, 550018, China
| | - Ziao Yuan
- College of Life Science and Technology, Harbin Normal University, Harbin, 150040, China
| | - Weiqi Zhang
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, 150040, China
| | - Jialong Wang
- Institute of Applied Microbiology, Heilongjiang Academy of Sciences, Harbin, 150010, China.
| | - Minghai Zhang
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, 150040, China.
| |
Collapse
|
4
|
Somers SE, Davidson GL, Mbandlwa P, McKeon CM, Stanton C, Ross RP, Quinn JL. Manipulating a host-native microbial strain compensates for low microbial diversity by increasing weight gain in a wild bird population. Proc Natl Acad Sci U S A 2024; 121:e2402352121. [PMID: 39401350 PMCID: PMC11513901 DOI: 10.1073/pnas.2402352121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 08/25/2024] [Indexed: 10/30/2024] Open
Abstract
Empirical studies from laboratory systems and humans show that the gut microbiota is linked to host health. Similar evidence for effects on traits linked to fitness in nature is rare, not least because experimentally manipulating the gut microbiota is challenging. We isolated, characterized, and cultured a bacterial strain, Lactobacillus kimchicus APC4233, directly from a wild bird (the great tit Parus major) and provided it as a self-administered dietary supplement. We assessed the impact of the treatment on the host microbiota community, on weight, and tested whether the treatment affected a previous result linking microbiota alpha diversity to weight in nestlings. The treatment dramatically increased L. kimchicus abundance in the gut microbiota and increased alpha diversity. This effect was strongest in the youngest birds, validating earlier findings pointing to a brief developmental window when the gut microbiota are most sensitive. In time-lagged models, nestling weight was higher in the treatment birds suggesting L. kimchicus may have probiotic potential. There was also a positive time-lagged relationship between diversity and weight in control birds but not in the treatment birds, suggesting L. kimchicus helped birds compensate for low alpha diversity. We discuss why ecological context is likely key when predicting impacts of the microbiome. The manipulation of the gut microbiota with a host native strain in this wild population provides direct evidence for the role of the microbiota in the ecology and evolution of natural populations.
Collapse
Affiliation(s)
- Shane E. Somers
- School of Biological, Earth and Environmental Sciences, Distillery Fields, University College Cork, CorkT23 TK30, Ireland
- APC Microbiome Ireland, University College Cork, CorkT12 YT20, Ireland
| | - Gabrielle L. Davidson
- School of Biological Sciences, University of East Anglia, NorwichNR4 7TU, United Kingdom
| | - Philiswa Mbandlwa
- APC Microbiome Ireland, University College Cork, CorkT12 YT20, Ireland
- Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, CorkP61 CK84, Ireland
| | - Caroline M. McKeon
- Environment and Marine Sciences, Agri-Food and Biosciences Institute, Northern IrelandBT9 5PX, United Kingdom
- Zoology Department, School of Natural Sciences, Trinity College Dublin, DublinD02 PN40, Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, CorkT12 YT20, Ireland
- Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, CorkP61 CK84, Ireland
| | - R. Paul Ross
- APC Microbiome Ireland, University College Cork, CorkT12 YT20, Ireland
- Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, CorkP61 CK84, Ireland
| | - John L. Quinn
- School of Biological, Earth and Environmental Sciences, Distillery Fields, University College Cork, CorkT23 TK30, Ireland
- Environmental Research Institute, University College Cork, CorkT23 XE10, Ireland
| |
Collapse
|
5
|
Bai J, Tang L, Bi Y, Li M. Multi-omics insights into the energy compensation of rumen microbiota of grazing yaks in cold season. Front Microbiol 2024; 15:1467841. [PMID: 39444681 PMCID: PMC11496799 DOI: 10.3389/fmicb.2024.1467841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024] Open
Abstract
Background The ability of yaks to adapt to the extreme environment of low temperatures and hypoxia at cold seasons on the Qinghai-Tibet Plateau (QTP) is related to the host genome; however, the convergent evolution of rumen microbiomes in host adaption is unknown. Methods Here, we conducted a multi-omics study on the rumen fluid of grazing yaks from warm (July) and cold (December) seasons on the QTP to evaluate the convergent evolution of rumen microbiomes in the adaptation of grazing yaks to cold-seasons environments. Results The results showed that grazing yaks at cold seasons had higher fibrolytic enzyme activities and volatile fatty acids (VFAs) concentrations, and the relative abundance of Firmicutes and the ratio Firmicutes to Bacteroidetes was significantly higher than that of yaks at warm seasons. Macrogenomic analyses showed that genes involved in forming VFAs and arginine were significantly enriched in cold-season yaks. Transcriptome analyses of the rumen epithelium showed that 72 genes associated with VFAs absorption and transport were significantly upregulated in cold-season yaks. Metabolomic analyses showed that the levels of ornithine, related to efficient nitrogen utilization, were significantly upregulated in cold-season yaks. Conclusion The synergistic role of rumen microbiomes in the adaptation of grazing yaks to extreme environments at cold seasons was revealed by multi-omics study.
Collapse
Affiliation(s)
- Jie Bai
- Key Laboratory for Grassland Ecosystem of Ministry of Education, College of Pratacultural Science, Gansu Agricultural University, Lanzhou, China
| | - Lijuan Tang
- Key Laboratory for Grassland Ecosystem of Ministry of Education, College of Pratacultural Science, Gansu Agricultural University, Lanzhou, China
| | - Yanliang Bi
- National Engineering Research Center of Biological Feed, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Mingliang Li
- Livestock and Poultry Genetic Resources Protection and Utilization Center in Qinghai Province, Xining, China
| |
Collapse
|
6
|
Li FXZ, Xu F, Li CC, Lei LM, Shan SK, Zheng MH, Lin X, Guo B, Tang KX, Duan JY, Wu YY, Cao YC, Liu JJ, Yuan LQ. Cold Exposure Alleviates T2DM Through Plasma-Derived Extracellular Vesicles. Int J Nanomedicine 2024; 19:10077-10095. [PMID: 39371478 PMCID: PMC11456273 DOI: 10.2147/ijn.s441847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 06/14/2024] [Indexed: 10/08/2024] Open
Abstract
Purpose Anecdotal reports have praised the benefits of cold exposure, exemplified by activities like winter swimming and cold water immersion. Cold exposure has garnered acclaim for its potential to confer benefits and potentially alleviate diabetes. We posited that systemic cold temperature (CT, 4-8°C) likely influences the organism's blood components through ambient temperature, prompting our investigation into the effects of chronic cold exposure on type 2 diabetic (T2DM) mice and our initial exploration of how cold exposure mitigates the incidence of T2DM. Methods The effects of CT (4-8°C) or room temperature (RT, 22-25°C) on T2DM mice were investigated. Mice blood and organ specimens were collected for fully automated biochemical testing, ELISA, HE staining, immunohistochemistry, and immunofluorescence. Glucose uptake was assessed using flow cytometry with 2-NBDG. Changes in potential signaling pathways such as protein kinase B (AKT), phosphorylated AKT (p-AKT), insulin receptor substrates 1 (IRS1), and phosphorylated IRS1 (p-IRS1) were evaluated by Western blot. Results CT or CT mice plasma-derived extracellular vesicles (CT-EVs) remarkably reduced blood glucose levels and improved insulin sensitivity in T2DM mice. This treatment enhanced glucose metabolism, systemic insulin sensitivity, and insulin secretion function while promoting glycogen accumulation in the liver and muscle. Additionally, CT-EVs treatment protected against the streptozocin (STZ)-induced destruction of islets in T2DM mice by inhibiting β-cell apoptosis. CT-EVs also shielded islets from destruction and increased the expression of p-IRS1 and p-AKT in adipocytes and hepatocytes. In vitro experiments further confirmed its pro-insulin sensitivity effect. Conclusion Our data indicate that cold exposure may have a potentially beneficial effect on the development of T2DM, mainly through the anti-diabetic effect of plasma-derived EVs released during cold stimulation. This phenomenon could significantly contribute to understanding the lower prevalence of diabetes in colder regions.
Collapse
Affiliation(s)
- Fu-Xing-Zi Li
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People’s Republic of China
| | - Feng Xu
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People’s Republic of China
| | - Chang-Chun Li
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People’s Republic of China
| | - Li-Min Lei
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People’s Republic of China
| | - Su-Kang Shan
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People’s Republic of China
| | - Ming-Hui Zheng
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People’s Republic of China
| | - Xiao Lin
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People’s Republic of China
| | - Bei Guo
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People’s Republic of China
| | - Ke-Xin Tang
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People’s Republic of China
| | - Jia-Yue Duan
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People’s Republic of China
| | - Yun-Yun Wu
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People’s Republic of China
| | - Ye-Chi Cao
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People’s Republic of China
| | - Jun-Jie Liu
- Department of Periodontal Division, Hunan Xiangya Stomatological Hospital, Central South University, Changsha, Hunan, 410008, People’s Republic of China
| | - Ling-Qing Yuan
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People’s Republic of China
| |
Collapse
|
7
|
Liukkonen M, Muriel J, Martínez-Padilla J, Nord A, Pakanen VM, Rosivall B, Tilgar V, van Oers K, Grond K, Ruuskanen S. Seasonal and environmental factors contribute to the variation in the gut microbiome: A large-scale study of a small bird. J Anim Ecol 2024; 93:1475-1492. [PMID: 39041321 DOI: 10.1111/1365-2656.14153] [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: 11/14/2023] [Accepted: 06/25/2024] [Indexed: 07/24/2024]
Abstract
Environmental variation can shape the gut microbiome, but broad/large-scale data on among and within-population heterogeneity in the gut microbiome and the associated environmental factors of wild populations is lacking. Furthermore, previous studies have limited taxonomical coverage, and knowledge about wild avian gut microbiomes is still scarce. We investigated large-scale environmental variation in the gut microbiome of wild adult great tits across the species' European distribution range. We collected fecal samples to represent the gut microbiome and used the 16S rRNA gene sequencing to characterize the bacterial gut microbiome. Our results show that gut microbiome diversity is higher during winter and that there are compositional differences between winter and summer gut microbiomes. During winter, individuals inhabiting mixed forest habitat show higher gut microbiome diversity, whereas there was no similar association during summer. Also, temperature was found to be a small contributor to compositional differences in the gut microbiome. We did not find significant differences in the gut microbiome among populations, nor any association between latitude, rainfall and the gut microbiome. The results suggest that there is a seasonal change in wild avian gut microbiomes, but that there are still many unknown factors that shape the gut microbiome of wild bird populations.
Collapse
Affiliation(s)
- Martta Liukkonen
- Department of Biological and Environmental Science, University of Jyväskylä, Jyväskylä, Finland
| | - Jaime Muriel
- Department of Biology, University of Turku, Turku, Finland
| | - Jesús Martínez-Padilla
- Department of Biodiversity Conservation and Ecosystem Restoration, Pyrenean Institute of Ecology (IPE-CSIC), Jaca, Spain
| | - Andreas Nord
- Department of Biology, Lund University, Lund, Sweden
| | | | - Balázs Rosivall
- Behavioural Ecology Group, Department of Systematic Zoology and Ecology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Vallo Tilgar
- Department of Zoology, Tartu University, Tartu, Estonia
| | - Kees van Oers
- Netherlands Institute of Ecology (NIOO-KNAW), Wageningen, The Netherlands
| | - Kirsten Grond
- Department of Biological Sciences, University of Alaska Anchorage, Anchorage, Alaska, USA
| | - Suvi Ruuskanen
- Department of Biological and Environmental Science, University of Jyväskylä, Jyväskylä, Finland
- Department of Biology, University of Turku, Turku, Finland
| |
Collapse
|
8
|
Chakraborty N, Hoke A, Campbell R, Holmes-Hampton G, Kumar VP, Moyler C, Gautam A, Hammamieh R, Ghosh SP. Ionizing Radiation Dose Differentially Affects the Host-Microbe Relationship over Time. Microorganisms 2024; 12:1995. [PMID: 39458305 PMCID: PMC11509422 DOI: 10.3390/microorganisms12101995] [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: 08/13/2024] [Revised: 09/05/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
Microorganisms that colonize in or on a host play significant roles in regulating the host's immunological fitness and bioenergy production, thus controlling the host's stress responses. Radiation elicits a pro-inflammatory and bioenergy-expensive state, which could influence the gut microbial compositions and, therefore, the host-microbe bidirectional relationship. To test this hypothesis, young adult mice were exposed to total body irradiation (TBI) at doses of 9.5 Gy and 11 Gy, respectively. The irradiated mice were euthanized on days 1, 3, and 9 post TBI, and their descending colon contents (DCCs) were collected. The 16S ribosomal RNAs from the DCCs were screened to find the differentially enriched bacterial taxa due to TBI. Subsequently, these data were analyzed to identify the metagenome-specific biofunctions. The bacterial community of the DCCs showed increased levels of diversity as time progressed following TBI. The abundance profile was the most divergent at day 9 post 11 Gy TBI. For instance, an anti-inflammatory and energy-harvesting bacterium, namely, Firmicutes, became highly abundant and co-expressed in the DCC with pro-inflammatory Deferribacteres at day 9 post 11 Gy TBI. A systems evaluation found a diverging trend in the regulation profiles of the functional networks that were linked to the bacteria and metabolites of the DCCs, respectively. Additionally, the network clusters associated with lipid metabolism and bioenergy synthesis were found to be activated in the DCC bacteria but inhibited in the metabolite space at day 9 post 11 Gy. Taking these results together, the present analysis indicated a disrupted mouse-bacteria symbiotic relationship as time progressed after lethal irradiation. This information can help develop precise interventions to ameliorate the symptoms triggered by TBI.
Collapse
Affiliation(s)
- Nabarun Chakraborty
- Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (N.C.); (A.H.); (R.C.); (C.M.); (A.G.)
| | - Allison Hoke
- Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (N.C.); (A.H.); (R.C.); (C.M.); (A.G.)
| | - Ross Campbell
- Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (N.C.); (A.H.); (R.C.); (C.M.); (A.G.)
- The Geneva Foundation, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Gregory Holmes-Hampton
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences (USUHS), Bethesda, MD 20889, USA; (G.H.-H.); (V.P.K.)
| | - Vidya P. Kumar
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences (USUHS), Bethesda, MD 20889, USA; (G.H.-H.); (V.P.K.)
| | - Candace Moyler
- Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (N.C.); (A.H.); (R.C.); (C.M.); (A.G.)
- The Geneva Foundation, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Aarti Gautam
- Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (N.C.); (A.H.); (R.C.); (C.M.); (A.G.)
| | - Rasha Hammamieh
- Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (N.C.); (A.H.); (R.C.); (C.M.); (A.G.)
| | - Sanchita P. Ghosh
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences (USUHS), Bethesda, MD 20889, USA; (G.H.-H.); (V.P.K.)
| |
Collapse
|
9
|
Li K, Zhang G, Sun M, Xia M, Shi R, Jin Y, Zhang X. Comparative Analysis of the Potential Adaptability of Tibetan Dzo and Yellow Cattle Based on Blood Indices, Metabolites, and Fecal Microbiota. Animals (Basel) 2024; 14:2728. [PMID: 39335317 PMCID: PMC11429423 DOI: 10.3390/ani14182728] [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: 08/13/2024] [Revised: 09/10/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
This study aimed to investigate the differences in environmental adaptability between dzo and Tibetan yellow cattle by using corresponding assay kits to analyze blood indices, utilizing mass spectrometry for blood metabolite profiling, and performing 16S rDNA sequencing of fecal microbiota. Forty female cattle were randomly divided into a dzomo (female dzo) group (MG, n = 20) and a Tibetan-yellow-cattle group (HG, n = 20). After 150 days of uniform feeding, six cattle from each group were randomly picked for jugular blood sampling and collection of fecal microorganisms. The results showed that the serum albumin, creatinine, total protein, superoxide dismutase, IgG, and IgM concentrations in the MG group were higher (p < 0.05), whereas the serum triglyceride concentration was lower, compared to the HG group (p < 0.05). The higher level of phospholipids containing long-chain polyunsaturated fatty acids (PUFAs) (PC (18:5e/2:0), PC (20:5e/2:0), LPC 18:2, LPC 20:5) observed in the serum of the dzo suggests that they have an advantage in adapting to the challenging conditions of the plateau environment. The fecal microbiota analysis showed that Akkermansia was significantly enriched in the MG group; this might be the key bacterial genus leading to the strong adaptability of dzo. Our findings indicated the dzo's superior adaptation to the Tibetan Plateau's harsh environment.
Collapse
Affiliation(s)
- Kenan Li
- Grassland Research Institute of Chinese Academy of Agricultural Sciences, Hohhot 010010, China
| | - Guorui Zhang
- Grassland Research Institute of Chinese Academy of Agricultural Sciences, Hohhot 010010, China
- College of Prataculture, Qingdao Agricultural University, Qingdao 266200, China
| | - Mengjiao Sun
- College of Prataculture, Qingdao Agricultural University, Qingdao 266200, China
| | - Maolin Xia
- Tibet Autonomous Region Animal Husbandry Station, Lhasa 850000, China
| | - Ruizhi Shi
- Institute of Practaculture Science, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa 850000, China
| | - Yanmei Jin
- Marine College, Shandong University, Weihai 264209, China
| | - Xiaoqing Zhang
- Grassland Research Institute of Chinese Academy of Agricultural Sciences, Hohhot 010010, China
- Institute of Practaculture Science, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa 850000, China
| |
Collapse
|
10
|
Duan T, Alim A, Tian H, Li T. Roundup-Induced Gut Dysbiosis, Irrelevant to Aromatic Amino Acid Deficiency, Impairs the Gut Function in Rats. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024. [PMID: 39302074 DOI: 10.1021/acs.jafc.4c04045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
Glyphosate, the most popular herbicide globally, has long been considered safe for mammals. However, whether glyphosate can disturb gut microbiota via inhibiting aromatic amino acid (AAA) synthesis has been under debate recently. Here, we evaluated the impacts of chronic exposure to Roundup on gut health with the addition of AAA and explored the mechanism behind Roundup-induced gut dysfunction by performing fecal microbiota transplantation. 500 mg/kg·bw of Roundup, independent of AAA deficiency, caused severe damage to gut function, as characterized by gut microbial dysbiosis, oxidative stress damage, intestinal inflammation, and histopathological injury, particularly in female rats. Notably, similar to Roundup, Roundup-shaped gut microbiome evidently damaged serum, cecum, and colon profiling of oxidative stress biomarkers (malonaldehyde (MDA), glutathione peroxidase (GSH-Px), superoxide dismutase (SOD), glutathione (GSH), and H2O2). Moreover, it induced 0.65-, 3.29-, and 2.36-fold increases in colonic IL-1β, IL-6, and TNF-α levels, and 0.34-fold decreases in the IL-10 level. Upon transplanting healthy fecal microbiota to Roundup-treated rats, they exhibited a healthier gut microenvironment with mitigated inflammation, oxidative damage, and intestinal injury. Overall, our findings provide new insights into the safety of Roundup, highlight the crucial role of gut microbiota in Roundup-induced gut dysfunction, and pave the way for developing gut-microbiota-based strategies to address Roundup-related safety issues.
Collapse
Affiliation(s)
- Tianchi Duan
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Aamina Alim
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Honglei Tian
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Ting Li
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| |
Collapse
|
11
|
Soldán M, Argalášová Ľ, Hadvinová L, Galileo B, Babjaková J. The Effect of Dietary Types on Gut Microbiota Composition and Development of Non-Communicable Diseases: A Narrative Review. Nutrients 2024; 16:3134. [PMID: 39339734 PMCID: PMC11434870 DOI: 10.3390/nu16183134] [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: 07/26/2024] [Revised: 09/14/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024] Open
Abstract
INTRODUCTION The importance of diet in shaping the gut microbiota is well established and may help improve an individual's overall health. Many other factors, such as genetics, age, exercise, antibiotic therapy, or tobacco use, also play a role in influencing gut microbiota. AIM This narrative review summarizes how three distinct dietary types (plant-based, Mediterranean, and Western) affect the composition of gut microbiota and the development of non-communicable diseases (NCDs). METHODS A comprehensive literature search was conducted using the PubMed, Web of Science, and Scopus databases, focusing on the keywords "dietary pattern", "gut microbiota" and "dysbiosis". RESULTS Both plant-based and Mediterranean diets have been shown to promote the production of beneficial bacterial metabolites, such as short-chain fatty acids (SCFAs), while simultaneously lowering concentrations of trimethylamine-N-oxide (TMAO), a molecule associated with negative health outcomes. Additionally, they have a positive impact on microbial diversity and therefore are generally considered healthy dietary types. On the other hand, the Western diet is a typical example of an unhealthy nutritional approach leading to an overgrowth of pathogenic bacteria, where TMAO levels rise and SCFA production drops due to gut dysbiosis. CONCLUSION The current scientific literature consistently highlights the superiority of plant-based and Mediterranean dietary types over the Western diet in promoting gut health and preventing NCDs. Understanding the influence of diet on gut microbiota modulation may pave the way for novel therapeutic strategies.
Collapse
Affiliation(s)
| | - Ľubica Argalášová
- Institute of Hygiene, Faculty of Medicine, Comenius University in Bratislava, Špitálska 24, 813 72 Bratislava, Slovakia; (M.S.); (L.H.); (B.G.); (J.B.)
| | | | | | | |
Collapse
|
12
|
Sun C, Su J, Wang J, Ding K, Chen C. Lycium barbarum polysaccharide increases thermogenesis and energy metabolism through modulation of the gut microbiota to confer resistance to cold temperatures. FASEB J 2024; 38:e70010. [PMID: 39230621 DOI: 10.1096/fj.202400870r] [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: 04/17/2024] [Revised: 07/18/2024] [Accepted: 08/09/2024] [Indexed: 09/05/2024]
Abstract
Traditional Chinese medical literature contains numerous records of many traditional Chinese herbal medicines that exhibit efficacy in enhancing resistance to cold, yet there is a lack of scientific explanation. Lycium barbarum is among the herbal medicines that are explicitly documented to enhance resistance to cold in the "Ben Cao Gang Mu (Compendium of Materia Medica)". Herein, we investigated L. barbarum polysaccharide (LBP)-induced browning of inguinal white adipose tissue (iWAT), energy expenditure and thermogenic function in a long-term (4 months) treatment mouse model. LBP supplementation resulted in a significant reduction in weight and adipocyte size in iWAT, along with increased gut microbiota diversity. Specifically, the levels of Lachnospiraceae, Ruminococcaceae and Bacteroidaceae (short-chain fatty acid-producing bacteria) were elevated, leading to a higher level of short-chain fatty acids (SCFAs) in the caecal content. These effects subsequently triggered the release of glucagon-like peptide-1 (GLP-1) and activated the CREB/PGC1α signaling pathway in iWAT, thereby increasing energy expenditure and enhancing thermogenic function. The antibiotic treatment experiments confirmed that the LBP-mediated gut microbiota participated in the process of iWAT browning. In summary, our findings provide the first scientific explanation and mechanistic insights into the cold resistance of L. barbarum and identify potentially safe natural product supplements for individuals in alpine areas.
Collapse
Affiliation(s)
- Chuanxin Sun
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, P.R. China
| | - Juan Su
- CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Jiarui Wang
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, P.R. China
- College of Life Science, University of Chinese Academy of Sciences, Beijing, P.R. China
| | - Kan Ding
- CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P.R. China
- College of Life Science, University of Chinese Academy of Sciences, Beijing, P.R. China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Science, SSIP Healthcare and Medicine Demonstration Zone, Zhongshan, Guangdong, P.R. China
| | - Chang Chen
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, P.R. China
- College of Life Science, University of Chinese Academy of Sciences, Beijing, P.R. China
- Beijing Institute for Brain Disorders Capital Medical University, Beijing, P.R. China
| |
Collapse
|
13
|
Chang Y, Zhang Z, Cai J, Wang C, Liu D, Liu Z, Xu C. Coevolution of specific gut microbiota of Min pig with host cold adaptation through enhanced vitamin B1 synthesis. Front Microbiol 2024; 15:1448090. [PMID: 39282562 PMCID: PMC11401075 DOI: 10.3389/fmicb.2024.1448090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 07/30/2024] [Indexed: 09/19/2024] Open
Abstract
Min pigs exhibit remarkable cold tolerance, where vitamin B1 synthesis by gut microbiota is crucial for the host's energy metabolism. However, the role of this synthesis in cold adaptation of Min pigs are not yet fully understood. This study utilized 16S rRNA amplicon and metagenomic sequencing to examine seasonal variations in the gut microbiota of Min pigs. Results indicated a significant rise in microbial diversity in winter, with the Bacteroidetes group being the most notably increased. The vitamin B1 biosynthetic pathway was significantly enriched during winter, with six significantly upregulated genes (ThiC, ThiD, ThiE, ThiG, ThiH, and ThiL) showing strong evidence of purifying selection. Among the six vitamin B1 synthesis genes significantly upregulated during winter, the increase was mainly due to a marked elevation in several sequences from specific microbial species. Binding energy analysis revealed that, except for ThiL, the average substrate binding energy of the top 10 sequences with the largest seasonal differences was significantly lower than those of the 10 sequences with the smallest differences. Furthermore, most of these sequences were uniquely prevalent in Min pigs and were not found in the homologous sequences of Duroc pigs. Bacteroidetes and Bacteroidales were identified as the primary contributors to these gene sequences. This research provides valuable insights for developing innovative cold-resistant feed and probiotics.
Collapse
Affiliation(s)
- Yang Chang
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin, China
- College of Life Science, Northeast Agricultural University, Harbin, China
| | - Ziwen Zhang
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin, China
- College of Life Science, Northeast Agricultural University, Harbin, China
| | | | | | - Di Liu
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin, China
| | - Zhonghua Liu
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin, China
- College of Life Science, Northeast Agricultural University, Harbin, China
| | - Chunzhu Xu
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin, China
- College of Life Science, Northeast Agricultural University, Harbin, China
| |
Collapse
|
14
|
Daubresse L, Portas A, Bertaud A, Marlinge M, Gaillard S, Risso JJ, Ramdani C, Rostain JC, Adjiriou N, Desruelle AV, Blatteau JE, Guieu R, Vallée N. CO 2 Breathing Prior to Simulated Diving Increases Decompression Sickness Risk in a Mouse Model: The Microbiota Trail Is Not Forgotten. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2024; 21:1141. [PMID: 39338024 PMCID: PMC11431549 DOI: 10.3390/ijerph21091141] [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: 07/05/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/30/2024]
Abstract
Decompression sickness (DCS) with neurological disorders is the leading cause of major diving accidents treated in hyperbaric chambers. Exposure to high levels of CO2 during diving is a safety concern for occupational groups at risk of DCS. However, the effects of prior exposure to CO2 have never been evaluated. The purpose of this study was to evaluate the effect of CO2 breathing prior to a provocative dive on the occurrence of DCS in mice. Fifty mice were exposed to a maximum CO2 concentration of 70 hPa, i.e., 7% at atmospheric pressure, for one hour at atmospheric pressure. Another 50 mice breathing air under similar conditions served as controls. In the AIR group (control), 22 out of 50 mice showed post-dive symptoms compared to 44 out of 50 in the CO2 group (p < 0.001). We found that CO2 breathing is associated with a decrease in body temperature in mice and that CO2 exposure dramatically increases the incidence of DCS (p < 0.001). More unexpectedly, it appears that the lower temperature of the animals even before exposure to the accident-prone protocol leads to an unfavorable prognosis (p = 0.046). This study also suggests that the composition of the microbiota may influence thermogenesis and thus accidentology. Depending on prior exposure, some of the bacterial genera identified in this work could be perceived as beneficial or pathogenic.
Collapse
Affiliation(s)
- Lucille Daubresse
- Service de Médecine Hyperbare, Hôpital d’Instruction des Armées, 83000 Toulon, France (J.-E.B.)
| | | | - Alexandrine Bertaud
- Aix-Marseille University, 27 Boulevard Jean-Moulin, 13005 Marseille, France (J.-C.R.); (N.A.); (R.G.)
| | - Marion Marlinge
- Aix-Marseille University, 27 Boulevard Jean-Moulin, 13005 Marseille, France (J.-C.R.); (N.A.); (R.G.)
| | | | - Jean-Jacques Risso
- Subaquatic Operational Research Team (ERRSO), Military Institute of Biomedical Research (IRBA), 83000 Toulon, France (C.R.); (A.-V.D.)
| | - Céline Ramdani
- Subaquatic Operational Research Team (ERRSO), Military Institute of Biomedical Research (IRBA), 83000 Toulon, France (C.R.); (A.-V.D.)
| | - Jean-Claude Rostain
- Aix-Marseille University, 27 Boulevard Jean-Moulin, 13005 Marseille, France (J.-C.R.); (N.A.); (R.G.)
| | - Nabil Adjiriou
- Aix-Marseille University, 27 Boulevard Jean-Moulin, 13005 Marseille, France (J.-C.R.); (N.A.); (R.G.)
| | - Anne-Virginie Desruelle
- Subaquatic Operational Research Team (ERRSO), Military Institute of Biomedical Research (IRBA), 83000 Toulon, France (C.R.); (A.-V.D.)
| | - Jean-Eric Blatteau
- Service de Médecine Hyperbare, Hôpital d’Instruction des Armées, 83000 Toulon, France (J.-E.B.)
| | - Régis Guieu
- Aix-Marseille University, 27 Boulevard Jean-Moulin, 13005 Marseille, France (J.-C.R.); (N.A.); (R.G.)
| | - Nicolas Vallée
- Subaquatic Operational Research Team (ERRSO), Military Institute of Biomedical Research (IRBA), 83000 Toulon, France (C.R.); (A.-V.D.)
| |
Collapse
|
15
|
Molica F, Ehrlich A, Pelli G, Rusiecka OM, Montessuit C, Chanson M, Kwak BR. Cold Exposure Rejuvenates the Metabolic Phenotype of Panx1-/- Mice. Biomolecules 2024; 14:1058. [PMID: 39334824 PMCID: PMC11430693 DOI: 10.3390/biom14091058] [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: 07/02/2024] [Revised: 08/15/2024] [Accepted: 08/23/2024] [Indexed: 09/30/2024] Open
Abstract
Pannexin1 (Panx1) ATP channels are important in adipocyte biology, potentially influencing energy storage and expenditure. We compared the metabolic phenotype of young (14 weeks old) and mature (20 weeks old) wild-type (WT) and Panx1-/- mice exposed or not to cold (6 °C) during 28 days, a condition promoting adipocyte browning. Young Panx1-/- mice weighed less and exhibited increased fat mass, improved glucose tolerance, and lower insulin sensitivity than WT mice. Their energy expenditure and respiratory exchange ratio (RER) were increased, and their fatty acid oxidation decreased. These metabolic effects were no longer observed in mature Panx1-/- mice. The exposure of mature mice to cold exacerbated their younger metabolic phenotype. The white adipose tissue (WAT) of cold-exposed Panx1-/- mice contained more small-sized adipocytes, but, in contrast to WT mice, white adipocytes did not increase their expression of Ucp1 nor of other markers of browning adipocytes. Interestingly, Glut4 expression was already enhanced in the WAT of young Panx1-/- mice kept at 22 °C as compared to WT mice. Thus, Panx1 deletion exerts overall beneficial metabolic effects in mice that are pre-adapted to chronic cold exposure. Panx1-/- mice show morphological characteristics of WAT browning, which are exacerbated upon cold exposure, an effect that appears to be associated with Ucp1-independent thermogenesis.
Collapse
Affiliation(s)
- Filippo Molica
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland; (A.E.); (G.P.); (O.M.R.); (C.M.); (B.R.K.)
- Geneva Center for Inflammation Research, CH-1211 Geneva, Switzerland;
| | - Avigail Ehrlich
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland; (A.E.); (G.P.); (O.M.R.); (C.M.); (B.R.K.)
- Geneva Center for Inflammation Research, CH-1211 Geneva, Switzerland;
| | - Graziano Pelli
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland; (A.E.); (G.P.); (O.M.R.); (C.M.); (B.R.K.)
- Geneva Center for Inflammation Research, CH-1211 Geneva, Switzerland;
| | - Olga M. Rusiecka
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland; (A.E.); (G.P.); (O.M.R.); (C.M.); (B.R.K.)
- Geneva Center for Inflammation Research, CH-1211 Geneva, Switzerland;
| | - Christophe Montessuit
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland; (A.E.); (G.P.); (O.M.R.); (C.M.); (B.R.K.)
| | - Marc Chanson
- Geneva Center for Inflammation Research, CH-1211 Geneva, Switzerland;
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Brenda R. Kwak
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland; (A.E.); (G.P.); (O.M.R.); (C.M.); (B.R.K.)
- Geneva Center for Inflammation Research, CH-1211 Geneva, Switzerland;
| |
Collapse
|
16
|
Tang X, Zhang L, Wang L, Ren S, Zhang J, Ma Y, Xu F, Wu G, Zhang Y. Multi-Omics Analysis Reveals Dietary Fiber's Impact on Growth, Slaughter Performance, and Gut Microbiome in Durco × Bamei Crossbred Pig. Microorganisms 2024; 12:1674. [PMID: 39203515 PMCID: PMC11357262 DOI: 10.3390/microorganisms12081674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 07/28/2024] [Accepted: 08/07/2024] [Indexed: 09/03/2024] Open
Abstract
Dietary fiber (DF) is an important nutrient component in pig's diet that remarkably influences their growth and slaughter performance. The ability of pigs to digest DF depends on the microbial composition of the intestinal tract, particularly in the hindgut. However, studies on how DF alters the growth and slaughter performance of pigs by shaping the gut microbial composition and metabolites are still limited. Therefore, this study aimed to investigate the effects of DF on microbial composition, functions, and metabolites, ultimately altering host growth and slaughter performance using Durco × Bamei crossbred pigs supplemented with 0%, 10%, 17%, and 24% broad bean silage in the basic diet. We found that the final weight, average daily gain, fat, and lean meat weight significantly decreased with increasing DF. Pigs with the lowest slaughter rate and fat weight were observed in the 24% fiber-supplemented group. Gut microbial communities with the highest alpha diversity were formed in the 17% fiber group. The relative abundance of fiber-degrading bacteria, bile acid, and succinate-producing bacteria, including Prevotella sp., Bacteroides sp., Ruminococcus sp., and Parabacteroides sp., and functional pathways, including the butanoate metabolism and the tricarboxylic acid [TCA] cycle, significantly increased in the high-fiber groups. The concentrations of several bile acids significantly decreased in the fiber-supplemented groups, whereas the concentrations of succinate and long-chain fatty acids increased. Our results indicate that a high-fiber diet may alter the growth and slaughter performance of Durco × Bamei crossbred pigs by modulating the composition of Prevotella sp., Bacteroides sp., Ruminococcus sp., Parabacteroides sp., and metabolite pathways of bile acids and succinate.
Collapse
Affiliation(s)
- Xianjiang Tang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, China
- Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining 810008, China
| | - Liangzhi Zhang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, China
- Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining 810008, China
| | - Lei Wang
- Plateau Livestock Genetic Resources Protection and Innovative Utilization Key Laboratory of Qinghai Province, Qinghai Academy of Animal and Veterinary Medicine, Qinghai University, Xining 810016, China
| | - Shien Ren
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, China
- Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining 810008, China
| | - Jianbo Zhang
- Plateau Livestock Genetic Resources Protection and Innovative Utilization Key Laboratory of Qinghai Province, Qinghai Academy of Animal and Veterinary Medicine, Qinghai University, Xining 810016, China
| | - Yuhong Ma
- Plateau Livestock Genetic Resources Protection and Innovative Utilization Key Laboratory of Qinghai Province, Qinghai Academy of Animal and Veterinary Medicine, Qinghai University, Xining 810016, China
| | - Fafang Xu
- Plateau Livestock Genetic Resources Protection and Innovative Utilization Key Laboratory of Qinghai Province, Qinghai Academy of Animal and Veterinary Medicine, Qinghai University, Xining 810016, China
| | - Guofang Wu
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, China
- Plateau Livestock Genetic Resources Protection and Innovative Utilization Key Laboratory of Qinghai Province, Qinghai Academy of Animal and Veterinary Medicine, Qinghai University, Xining 810016, China
| | - Yanming Zhang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, China
- Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining 810008, China
| |
Collapse
|
17
|
Aagaard A, Bechsgaard J, Sørensen JG, Sandfeld T, Settepani V, Bird TL, Lund MB, Malmos KG, Falck-Rasmussen K, Darolti I, Nielsen KL, Johannsen M, Vosegaard T, Tregenza T, Verhoeven KJF, Mank JE, Schramm A, Bilde T. Molecular Mechanisms of Temperature Tolerance Plasticity in an Arthropod. Genome Biol Evol 2024; 16:evae165. [PMID: 39058286 DOI: 10.1093/gbe/evae165] [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: 05/28/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
How species thrive in a wide range of environments is a major focus of evolutionary biology. For many species, limited genetic diversity or gene flow among habitats means that phenotypic plasticity must play an important role in their capacity to tolerate environmental heterogeneity and to colonize new habitats. However, we have a limited understanding of the molecular components that govern plasticity in ecologically relevant phenotypes. We examined this hypothesis in a spider species (Stegodyphus dumicola) with extremely low species-wide genetic diversity that nevertheless occupies a broad range of thermal environments. We determined phenotypic responses to temperature stress in individuals from four climatic zones using common garden acclimation experiments to disentangle phenotypic plasticity from genetic adaptations. Simultaneously, we created data sets on multiple molecular modalities: the genome, the transcriptome, the methylome, the metabolome, and the bacterial microbiome to determine associations with phenotypic responses. Analyses of phenotypic and molecular associations reveal that acclimation responses in the transcriptome and metabolome correlate with patterns of phenotypic plasticity in temperature tolerance. Surprisingly, genes whose expression seemed to be involved in plasticity in temperature tolerance were generally highly methylated contradicting the idea that DNA methylation stabilizes gene expression. This suggests that the function of DNA methylation in invertebrates varies not only among species but also among genes. The bacterial microbiome was stable across the acclimation period; combined with our previous demonstrations that the microbiome is temporally stable in wild populations, this is convincing evidence that the microbiome does not facilitate plasticity in temperature tolerance. Our results suggest that population-specific variation in temperature tolerance among acclimation temperatures appears to result from the evolution of plasticity in mainly gene expression.
Collapse
Affiliation(s)
- Anne Aagaard
- Section for Genetics, Ecology and Evolution, Centre for EcoGenetics, Department of Biology, Aarhus University, Aarhus C, Denmark
| | - Jesper Bechsgaard
- Section for Genetics, Ecology and Evolution, Centre for EcoGenetics, Department of Biology, Aarhus University, Aarhus C, Denmark
| | - Jesper Givskov Sørensen
- Section for Genetics, Ecology and Evolution, Centre for EcoGenetics, Department of Biology, Aarhus University, Aarhus C, Denmark
| | - Tobias Sandfeld
- Section for Microbiology, Department of Biology, Aarhus University, Aarhus C, Denmark
| | - Virginia Settepani
- Section for Genetics, Ecology and Evolution, Centre for EcoGenetics, Department of Biology, Aarhus University, Aarhus C, Denmark
| | - Tharina L Bird
- General Entomology, DITSONG: National Museum of Natural History, Pretoria, South Africa
- Department of Zoology and Entomology, University of Pretoria, Pretoria, South Africa
- Department of Arachnology and Myriapodology, National Museum of Namibia, Windhoek, Namibia
| | - Marie Braad Lund
- Section for Microbiology, Department of Biology, Aarhus University, Aarhus C, Denmark
| | - Kirsten Gade Malmos
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C, Denmark
| | - Kasper Falck-Rasmussen
- Section for Genetics, Ecology and Evolution, Centre for EcoGenetics, Department of Biology, Aarhus University, Aarhus C, Denmark
| | - Iulia Darolti
- Department of Zoology and Biodiversity Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Ecology and Evolution, University of Lausanne, Lausanne, Switzerland
| | | | - Mogens Johannsen
- Department of Forensic Medicine, Aarhus University, Aarhus N, Denmark
| | - Thomas Vosegaard
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C, Denmark
- Department of Chemistry, Aarhus University, Aarhus C, Denmark
| | - Tom Tregenza
- Centre for Ecology and Conservation, University of Exeter, Penryn Campus, Penryn TR109FE, UK
| | - Koen J F Verhoeven
- Terrestrial Ecology Department, Netherlands Institute of Ecology (NIOO-KNAW), Wageningen 6708 PB, The Netherlands
| | - Judith E Mank
- Department of Zoology and Biodiversity Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Andreas Schramm
- Section for Microbiology, Department of Biology, Aarhus University, Aarhus C, Denmark
| | - Trine Bilde
- Section for Genetics, Ecology and Evolution, Centre for EcoGenetics, Department of Biology, Aarhus University, Aarhus C, Denmark
- Centre for Ecology and Conservation, University of Exeter, Penryn Campus, Penryn TR109FE, UK
| |
Collapse
|
18
|
Lee J, Reiman D, Singh S, Chang A, Morel L, Chervonsky AV. Microbial influences on severity and sex bias of systemic autoimmunity. Immunol Rev 2024; 325:64-76. [PMID: 38716867 PMCID: PMC11338725 DOI: 10.1111/imr.13341] [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: 06/13/2024]
Abstract
Commensal microbes have the capacity to affect development and severity of autoimmune diseases. Germ-free (GF) animals have proven to be a fine tool to obtain definitive answers to the queries about the microbial role in these diseases. Moreover, GF and gnotobiotic animals can be used to dissect the complex symptoms and determine which are regulated (enhanced or attenuated) by microbes. These include disease manifestations that are sex biased. Here, we review comparative analyses conducted between GF and Specific-Pathogen Free (SPF) mouse models of autoimmunity. We present data from the B6;NZM-Sle1NZM2410/AegSle2NZM2410/AegSle3NZM2410/Aeg-/LmoJ (B6.NZM) mouse model of systemic lupus erythematosus (SLE) characterized by multiple measurable features. We compared the severity and sex bias of SPF, GF, and ex-GF mice and found variability in the severity and sex bias of some manifestations. Colonization of GF mice with the microbiotas taken from B6.NZM mice housed in two independent institutions variably affected severity and sexual dimorphism of different parameters. Thus, microbes regulate both the severity and sexual dimorphism of select SLE traits. The sensitivity of particular trait to microbial influence can be used to further dissect the mechanisms driving the disease. Our results demonstrate the complexity of the problem and open avenues for further investigations.
Collapse
Affiliation(s)
- Jean Lee
- Committee on Cancer Biology, The University of Chicago, Chicago, Illinois, USA
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
| | - Derek Reiman
- Toyota Technological Institute at Chicago, Chicago, Illinois, USA
| | - Samara Singh
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
| | - Anthony Chang
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
| | - Laurence Morel
- Department of Microbiology and Immunology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Alexander V Chervonsky
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
- Committee on Immunology, The University of Chicago, Chicago, Illinois, USA
- Committee on Microbiology, The University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
19
|
Ren Y, Huang P, Zhang L, Tang YF, Luo SL, She Z, Peng H, Chen YQ, Luo JW, Duan WX, Liu LJ, Liu LQ. Dual Regulation Mechanism of Obesity: DNA Methylation and Intestinal Flora. Biomedicines 2024; 12:1633. [PMID: 39200098 PMCID: PMC11351752 DOI: 10.3390/biomedicines12081633] [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: 06/12/2024] [Revised: 07/15/2024] [Accepted: 07/18/2024] [Indexed: 09/01/2024] Open
Abstract
Obesity is a multifactorial chronic inflammatory metabolic disorder, with pathogenesis influenced by genetic and non-genetic factors such as environment and diet. Intestinal microbes and their metabolites play significant roles in the occurrence and development of obesity by regulating energy metabolism, inducing chronic inflammation, and impacting intestinal hormone secretion. Epigenetics, which involves the regulation of host gene expression without changing the nucleotide sequence, provides an exact direction for us to understand how the environment, lifestyle factors, and other risk factors contribute to obesity. DNA methylation, as the most common epigenetic modification, is involved in the pathogenesis of various metabolic diseases. The epigenetic modification of the host is induced or regulated by the intestinal microbiota and their metabolites, linking the dynamic interaction between the microbiota and the host genome. In this review, we examined recent advancements in research, focusing on the involvement of intestinal microbiota and DNA methylation in the etiology and progression of obesity, as well as potential interactions between the two factors, providing novel perspectives and avenues for further elucidating the pathogenesis, prevention, and treatment of obesity.
Collapse
Affiliation(s)
- Yi Ren
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
- Department of Pediatrics, Haikou Hospital of the Maternal and Child Health, Haikou 570100, China
- Department of Children’s Healthcare, Hainan Modern Women and Children’s Medical, Haikou 570100, China
| | - Peng Huang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Lu Zhang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Yu-Fen Tang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Sen-Lin Luo
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Zhou She
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Hong Peng
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Yu-Qiong Chen
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Jin-Wen Luo
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Wang-Xin Duan
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Ling-Juan Liu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Li-Qun Liu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| |
Collapse
|
20
|
Zhang Y, Gao Y, Li C, Zhang YA, Lu Y, Ye J, Liu X. Parabacteroides distasonis regulates the infectivity and pathogenicity of SVCV at different water temperatures. MICROBIOME 2024; 12:128. [PMID: 39020382 PMCID: PMC11253412 DOI: 10.1186/s40168-024-01799-9] [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: 07/07/2023] [Accepted: 03/24/2024] [Indexed: 07/19/2024]
Abstract
BACKGROUND Spring viremia of carp virus (SVCV) infects a wide range of fish species and causes high mortality rates in aquaculture. This viral infection is characterized by seasonal outbreaks that are temperature-dependent. However, the specific mechanism behind temperature-dependent SVCV infectivity and pathogenicity remains unclear. Given the high sensitivity of the composition of intestinal microbiota to temperature changes, it would be interesting to investigate if the intestinal microbiota of fish could play a role in modulating the infectivity of SVCV at different temperatures. RESULTS Our study found that significantly higher infectivity and pathogenicity of SVCV infection in zebrafish occurred at relatively lower temperature. Comparative analysis of the intestinal microbiota in zebrafish exposed to high- and low-temperature conditions revealed that temperature influenced the abundance and diversity of the intestinal microbiota in zebrafish. A significantly higher abundance of Parabacteroides distasonis and its metabolite secondary bile acid (deoxycholic acid, DCA) was detected in the intestine of zebrafish exposed to high temperature. Both colonization of Parabacteroides distasonis and feeding of DCA to zebrafish at low temperature significantly reduced the mortality caused by SVCV. An in vitro assay demonstrated that DCA could inhibit the assembly and release of SVCV. Notably, DCA also showed an inhibitory effect on the infectious hematopoietic necrosis virus, another Rhabdoviridae member known to be more infectious at low temperature. CONCLUSIONS This study provides evidence that temperature can be an important factor to influence the composition of intestinal microbiota in zebrafish, consequently impacting the infectivity and pathogenicity of SVCV. The findings highlight the enrichment of Parabacteroides distasonis and its derivative, DCA, in the intestines of zebrafish raised at high temperature, and they possess an important role in preventing the infection of SVCV and other Rhabdoviridae members in host fish. Video Abstract.
Collapse
Affiliation(s)
- Yujun Zhang
- National Key Laboratory of Agricultural Microbiology, College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei, China
- Hubei Engineering Technology Research Center for Aquatic Animal Diseases Control and Prevention, Wuhan, Hubei, China
| | - Yan Gao
- National Key Laboratory of Agricultural Microbiology, College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei, China
- Hubei Engineering Technology Research Center for Aquatic Animal Diseases Control and Prevention, Wuhan, Hubei, China
- Ocean College, Hebei Agricultural University, Qinhuangdao, Hebei, China
| | - Chen Li
- National Key Laboratory of Agricultural Microbiology, College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei, China
- Hubei Engineering Technology Research Center for Aquatic Animal Diseases Control and Prevention, Wuhan, Hubei, China
| | - Yong-An Zhang
- National Key Laboratory of Agricultural Microbiology, College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei, China
- Hubei Engineering Technology Research Center for Aquatic Animal Diseases Control and Prevention, Wuhan, Hubei, China
| | - Yuanan Lu
- Department of Public Health Sciences, Thompson School of Social Work & Public Health, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Jing Ye
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China.
| | - Xueqin Liu
- National Key Laboratory of Agricultural Microbiology, College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei, China.
- Hubei Engineering Technology Research Center for Aquatic Animal Diseases Control and Prevention, Wuhan, Hubei, China.
| |
Collapse
|
21
|
Gao J, Zhang M, Zhang L, Wang N, Zhao Y, Ren D, Yang X. Dietary Pectin from Premna microphylla Turcz Leaves Prevents Obesity by Regulating Gut Microbiota and Lipid Metabolism in Mice Fed High-Fat Diet. Foods 2024; 13:2248. [PMID: 39063332 PMCID: PMC11275460 DOI: 10.3390/foods13142248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/27/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
The present study was designed to investigate the protective effects of pectin extracted from Premna microphylla Turcz leaves (PTP) against high-fat-diet (HFD)-induced lipid metabolism disorders and gut microbiota dysbiosis in obese mice. PTP was made using the acid extraction method, and it was found to be an acidic pectin that had relative mole percentages of 32.1%, 29.2%, and 26.2% for galacturonic acid, arabinose, and galactose, respectively. The administration of PTP in C57BL/6J mice inhibited the HFD-induced abnormal weight gain, visceral obesity, and dyslipidemia, and also improved insulin sensitivity, as revealed by the improved insulin tolerance and the decreased glucose levels during an insulin sensitivity test. These effects were linked to increased energy expenditure, as demonstrated by the upregulation of thermogenesis-related protein UCP1 expression in the brown adipose tissue (BAT) of PTP-treated mice. 16S rRNA gene sequencing revealed that PTP dramatically improved the HFD-induced gut dysbiosis by lowering the ratio of Firmicutes to Bacteroidetes and the quantity of potentially harmful bacteria. These findings may provide a theoretical basis for us to understand the functions and usages of PTP in alleviating obesity.
Collapse
Affiliation(s)
- Jiaobei Gao
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi’an 710119, China; (J.G.); (D.R.)
| | - Mengxue Zhang
- Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi’an 710119, China; (M.Z.); (L.Z.); (N.W.); (Y.Z.)
| | - Li Zhang
- Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi’an 710119, China; (M.Z.); (L.Z.); (N.W.); (Y.Z.)
| | - Nan Wang
- Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi’an 710119, China; (M.Z.); (L.Z.); (N.W.); (Y.Z.)
| | - Yan Zhao
- Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi’an 710119, China; (M.Z.); (L.Z.); (N.W.); (Y.Z.)
| | - Daoyuan Ren
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi’an 710119, China; (J.G.); (D.R.)
| | - Xingbin Yang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi’an 710119, China; (J.G.); (D.R.)
| |
Collapse
|
22
|
Wang Z, Song B, Yao J, Li X, Zhang Y, Tang Z, Yi G. Whole-genome analysis reveals distinct adaptation signatures to diverse environments in Chinese domestic pigs. J Anim Sci Biotechnol 2024; 15:97. [PMID: 38982489 PMCID: PMC11234542 DOI: 10.1186/s40104-024-01053-0] [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: 02/07/2024] [Accepted: 05/20/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND Long-term natural and artificial selection has resulted in many genetic footprints within the genomes of pig breeds across distinct agroecological zones. Nevertheless, the mechanisms by which these signatures contribute to phenotypic diversity and facilitate environmental adaptation remain unclear. RESULTS Here, we leveraged whole-genome sequencing data from 82 individuals from 6 domestic pig breeds originating in tropical, high-altitude, and frigid regions. Population genetic analysis suggested that habitat isolation significantly shaped the genetic diversity and contributed to population stratification in local Chinese pig breeds. Analysis of selection signals revealed regions under selection for adaptation in tropical (55.5 Mb), high-altitude (43.6 Mb), and frigid (17.72 Mb) regions. The potential functions of the selective sweep regions were linked to certain complex traits that might play critical roles in different geographic environments, including fat coverage in frigid environments and blood indicators in tropical and high-altitude environments. Candidate genes under selection were significantly enriched in biological pathways involved in environmental adaptation. These pathways included blood circulation, protein degradation, and inflammation for adaptation to tropical environments; heart and lung development, hypoxia response, and DNA damage repair for high-altitude adaptation; and thermogenesis, cold-induced vasodilation (CIVD), and the cell cycle for adaptation to frigid environments. By examining the chromatin state of the selection signatures, we identified the lung and ileum as two candidate functional tissues for environmental adaptation. Finally, we identified a mutation (chr1: G246,175,129A) in the cis-regulatory region of ABCA1 as a plausible promising variant for adaptation to tropical environments. CONCLUSIONS In this study, we conducted a genome-wide exploration of the genetic mechanisms underlying the adaptability of local Chinese pig breeds to tropical, high-altitude, and frigid environments. Our findings shed light on the prominent role of cis-regulatory elements in environmental adaptation in pigs and may serve as a valuable biological model of human plateau-related disorders and cardiovascular diseases.
Collapse
Affiliation(s)
- Zhen Wang
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, China
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Foshan, 528226, China
| | - Bangmin Song
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, China
- School of Life Sciences, Henan University, Kaifeng, 475004, China
- Shenzhen Research Institute of Henan University, Shenzhen, 518000, China
| | - Jianyu Yao
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
| | - Xingzheng Li
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, China
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Foshan, 528226, China
| | - Yan Zhang
- Key Laboratory of Tropical Animal Breeding and Disease Research, Institute of Animal Science and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, 571100, China
| | - Zhonglin Tang
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, China.
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Foshan, 528226, China.
- Bama Yao Autonomous County Rural Revitalization Research Institute, Bama, 547500, China.
| | - Guoqiang Yi
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, China.
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Foshan, 528226, China.
- Bama Yao Autonomous County Rural Revitalization Research Institute, Bama, 547500, China.
| |
Collapse
|
23
|
Weng H, Deng L, Wang T, Xu H, Wu J, Zhou Q, Yu L, Chen B, Huang L, Qu Y, Zhou L, Chen X. Humid heat environment causes anxiety-like disorder via impairing gut microbiota and bile acid metabolism in mice. Nat Commun 2024; 15:5697. [PMID: 38972900 PMCID: PMC11228019 DOI: 10.1038/s41467-024-49972-w] [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: 11/11/2023] [Accepted: 06/25/2024] [Indexed: 07/09/2024] Open
Abstract
Climate and environmental changes threaten human mental health, but the impacts of specific environmental conditions on neuropsychiatric disorders remain largely unclear. Here, we show the impact of a humid heat environment on the brain and the gut microbiota using a conditioned housing male mouse model. We demonstrate that a humid heat environment can cause anxiety-like behaviour in male mice. Microbial 16 S rRNA sequencing analysis reveals that a humid heat environment caused gut microbiota dysbiosis (e.g., decreased abundance of Lactobacillus murinus), and metabolomics reveals an increase in serum levels of secondary bile acids (e.g., lithocholic acid). Moreover, increased neuroinflammation is indicated by the elevated expression of proinflammatory cytokines in the serum and cortex, activated PI3K/AKT/NF-κB signalling and a microglial response in the cortex. Strikingly, transplantation of the microbiota from mice reared in a humid heat environment readily recapitulates these abnormalities in germ-free mice, and these abnormalities are markedly reversed by Lactobacillus murinus administration. Human samples collected during the humid heat season also show a decrease in Lactobacillus murinus abundance and an increase in the serum lithocholic acid concentration. In conclusion, gut microbiota dysbiosis induced by a humid heat environment drives the progression of anxiety disorders by impairing bile acid metabolism and enhancing neuroinflammation, and probiotic administration is a potential therapeutic strategy for these disorders.
Collapse
Affiliation(s)
- Huandi Weng
- Department of Neurology and Stroke Center, The First Affiliated Hospital & Clinical Neuroscience Institute of Jinan University, Guangzhou, 510632, PR China
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, PR China
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, PR China
| | - Li Deng
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, PR China
| | - Tianyuan Wang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, PR China
| | - Huachong Xu
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, PR China
| | - Jialin Wu
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, PR China
| | - Qinji Zhou
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, PR China
| | - Lingtai Yu
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, PR China
| | - Boli Chen
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, PR China
| | - Li'an Huang
- Department of Neurology and Stroke Center, The First Affiliated Hospital & Clinical Neuroscience Institute of Jinan University, Guangzhou, 510632, PR China
| | - Yibo Qu
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, PR China
| | - Libing Zhou
- Department of Neurology and Stroke Center, The First Affiliated Hospital & Clinical Neuroscience Institute of Jinan University, Guangzhou, 510632, PR China.
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, PR China.
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, PR China.
- Neuroscience and Neurorehabilitation Institute, University of Health and Rehabilitation Sciences, Qingdao, 266071, Shandong, PR China.
- Center for Exercise and Brain Science, School of Psychology, Shanghai University of Sport, Shanghai, 200438, PR China.
| | - Xiaoyin Chen
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, PR China.
| |
Collapse
|
24
|
Vernetti A, Butler M, Banarjee C, Boxberger A, All K, Macari S, Chawarska K. Face-to-face live eye-tracking in toddlers with autism: Feasibility and impact of familiarity and face covering. Autism Res 2024; 17:1381-1390. [PMID: 38009948 DOI: 10.1002/aur.3060] [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: 06/14/2023] [Accepted: 10/31/2023] [Indexed: 11/29/2023]
Abstract
Studies utilizing eye-tracking methods have potential to promptly capture real-world dynamics of one of the core areas of vulnerability in autism spectrum disorders (ASD), selective social attention. So far, no studies have successfully reported utilizing the method to examine social attention in toddlers with neurodevelopmental vulnerabilities in real world and challenging settings such as an interactive face-to-face. This study examined the feasibility and validity of live eye-tracking method in response to live interaction occurring in several contexts in toddlers with and without ASD. Forty-seven toddlers with ASD, with atypical development (ATYP), or typically developing (TD), underwent a 30-s live eye-tracking procedure during a face-to-face interaction with a masked stranger using child-directed-speech (16 ASD, 14 ATYP, 17 TD; Mage = 23.44 months, SD = 6.02). Out of this group of toddlers, 29 (10 ASD, 8 ATYP, 11 TD, Mage = 21.97 months, SD = 5.76) underwent the same procedure with one of their maskless parent. Task completion rate, calibration accuracy, and affective response (feasibility measures) as well as attention to the task and the social partner (validity measures) were examined. Task completion rate and calibration accuracy were excellent. Despite the challenging context of face-to-face interaction, the toddlers exhibited a neutral affect, and high attention to the task and the speaker. As anticipated, toddlers with ASD looked less at the social partner compared with control groups. However, attention was comparable between the Stranger and Parent conditions, indicating that the effect was consistent regardless of presence of face covering or the familiarity of the interactive partner. The study demonstrates the high feasibility and validity of a live eye-tracking task involving face-to-face interaction in neurodiverse toddlers with social vulnerabilities. The effect of diminished attention to social partners in toddlers with autism is robust and present when interacting with an unfamiliar person and parent. The results suggest that a brief live eye-tracking method constitutes a promising ecologically valid candidate biomarker and potential intervention outcome in autism.
Collapse
Affiliation(s)
- Angelina Vernetti
- Child Study Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Maureen Butler
- Child Study Center, Northwestern University, Chicago, Illinois, USA
| | - Chitra Banarjee
- Child Study Center, University of Central Florida, Orlando, Florida, USA
| | - Alexandra Boxberger
- Child Study Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Katherine All
- Child Study Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Suzanne Macari
- Child Study Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Katarzyna Chawarska
- Child Study Center, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
25
|
Rana S, Canfield JR, Ward CS, Sprague JE. Bile acids and the gut microbiome are involved in the hyperthermia mediated by 3,4-methylenedioxymethamphetamine (MDMA). Sci Rep 2024; 14:14485. [PMID: 38914648 PMCID: PMC11196659 DOI: 10.1038/s41598-024-65433-2] [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: 02/02/2024] [Accepted: 06/20/2024] [Indexed: 06/26/2024] Open
Abstract
Hyperthermia induced by phenethylamines, such as 3,4-methylenedioxymethamphetamine (MDMA), can lead to life-threatening complications and death. Activation of the sympathetic nervous system and subsequent release of norepinephrine and activation of uncoupling proteins have been demonstrated to be the key mediators of phenethylamine-induced hyperthermia (PIH). Recently, the gut microbiome was shown to also play a contributing role in PIH. Here, the hypothesis that bile acids (BAs) produced by the gut microbiome are essential to PIH was tested. Changes in the serum concentrations of unconjugated primary BAs cholic acid (CA) and chenodeoxycholic acid (CDCA) and secondary BA deoxycholic acid (DCA) were measured following MDMA (20 mg/kg, sc) treatment in antibiotic treated and control rats. MDMA-induced a significant hyperthermic response and reduced the serum concentrations of three BAs 60 min post-treatment. Pretreatment with antibiotics (vancomycin, bacitracin and neomycin) in the drinking water for five days resulted in the depletion of BAs and a hypothermic response to MDMA. Gut bacterial communities in the antibiotic-treated group were distinct from the MDMA or saline treatment groups, with decreased microbiome diversity and alteration in taxa. Metagenomic functions inferred using the bioinformatic tool PICRUSt2 on 16S rRNA gene sequences indicated that bacterial genes associated to BA metabolism are less abundant in the antibiotic-MDMA treated group. Overall, these findings suggest that gut bacterial produced BAs might play an important role in MDMA-induced hyperthermia.
Collapse
Affiliation(s)
- Srishti Rana
- Department of Biological Sciences, Bowling Green State University, Bowling Green, OH, 43403, USA
| | - Jeremy R Canfield
- The Ohio Attorney General's Center for the Future of Forensic Science, Bowling Green State University, Bowling Green, OH, 43403, USA
| | - Christopher S Ward
- Department of Biological Sciences, Bowling Green State University, Bowling Green, OH, 43403, USA
| | - Jon E Sprague
- The Ohio Attorney General's Center for the Future of Forensic Science, Bowling Green State University, Bowling Green, OH, 43403, USA.
| |
Collapse
|
26
|
Lei LM, Li FXZ, Lin X, Xu F, Shan SK, Guo B, Zheng MH, Tang KX, Wang Y, Xu QS, Ouyang WL, Duan JY, Wu YY, Cao YC, Zhou ZA, He SY, Wu YL, Chen X, Lin ZJ, Pan Y, Yuan LQ, Li ZH. Cold exposure-induced plasma exosomes impair bone mass by inhibiting autophagy. J Nanobiotechnology 2024; 22:361. [PMID: 38910236 PMCID: PMC11194967 DOI: 10.1186/s12951-024-02640-z] [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: 12/19/2023] [Accepted: 06/14/2024] [Indexed: 06/25/2024] Open
Abstract
Recently, environmental temperature has been shown to regulate bone homeostasis. However, the mechanisms by which cold exposure affects bone mass remain unclear. In our present study, we observed that exposure to cold temperature (CT) decreased bone mass and quality in mice. Furthermore, a transplant of exosomes derived from the plasma of mice exposed to cold temperature (CT-EXO) can also impair the osteogenic differentiation of BMSCs and decrease bone mass by inhibiting autophagic activity. Rapamycin, a potent inducer of autophagy, can reverse cold exposure or CT-EXO-induced bone loss. Microarray sequencing revealed that cold exposure increases the miR-25-3p level in CT-EXO. Mechanistic studies showed that miR-25-3p can inhibit the osteogenic differentiation and autophagic activity of BMSCs. It is shown that inhibition of exosomes release or downregulation of miR-25-3p level can suppress CT-induced bone loss. This study identifies that CT-EXO mediates CT-induced osteoporotic effects through miR-25-3p by inhibiting autophagy via targeting SATB2, presenting a novel mechanism underlying the effect of cold temperature on bone mass.
Collapse
Affiliation(s)
- Li-Min Lei
- National Clinical Research Center for Metabolic Disease, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Fu-Xing-Zi Li
- National Clinical Research Center for Metabolic Disease, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiao Lin
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Feng Xu
- National Clinical Research Center for Metabolic Disease, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Su-Kang Shan
- National Clinical Research Center for Metabolic Disease, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Bei Guo
- National Clinical Research Center for Metabolic Disease, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ming-Hui Zheng
- National Clinical Research Center for Metabolic Disease, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ke-Xin Tang
- National Clinical Research Center for Metabolic Disease, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yi Wang
- National Clinical Research Center for Metabolic Disease, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qiu-Shuang Xu
- National Clinical Research Center for Metabolic Disease, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wen-Lu Ouyang
- National Clinical Research Center for Metabolic Disease, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jia-Yue Duan
- National Clinical Research Center for Metabolic Disease, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yun-Yun Wu
- National Clinical Research Center for Metabolic Disease, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ye-Chi Cao
- National Clinical Research Center for Metabolic Disease, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhi-Ang Zhou
- Department of Cardiovascular Surgery, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Si-Yang He
- National Clinical Research Center for Metabolic Disease, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yan-Lin Wu
- National Clinical Research Center for Metabolic Disease, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xi Chen
- National Clinical Research Center for Metabolic Disease, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zheng-Jun Lin
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Yi Pan
- Department of Endocrinology, The Second Affiliated Hospital of Kunming Medical University, No. 374 The Dianmian Avenue, Wuhua, Kunming, Yunnan, 650101, China
| | - Ling-Qing Yuan
- National Clinical Research Center for Metabolic Disease, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China.
| | - Zhi-Hong Li
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
- Department of Orthopaedics, Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
27
|
Álvarez-Herms J, Odriozola A. Microbiome and physical activity. ADVANCES IN GENETICS 2024; 111:409-450. [PMID: 38908903 DOI: 10.1016/bs.adgen.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/24/2024]
Abstract
Regular physical activity promotes health benefits and contributes to develop the individual biological potential. Chronical physical activity performed at moderate and high-intensity is the intensity more favorable to produce health development in athletes and improve the gut microbiota balance. The athletic microbiome is characterized by increased microbial diversity and abundance as well as greater phenotypic versatility. In addition, physical activity and microbiota composition have bidirectional effects, with regular physical activity improving microbial composition and microbial composition enhancing physical performance. The improvement of physical performance by a healthy microbiota is related to different phenotypes: i) efficient metabolic development, ii) improved regulation of intestinal permeability, iii) favourable modulation of local and systemic inflammatory and efficient immune responses, iv) efective regulation of systemic pH and, v) protection against acute stressful events such as environmental exposure to altitude or heat. The type of sport, both intensity or volume characteristics promote microbiota specialisation. Individual assessment of the state of the gut microbiota can be an effective biomarker for monitoring health in the medium to long term. The relationship between the microbiota and the rest of the body is bidirectional and symbiotic, with a full connection between the systemic functions of the nervous, musculoskeletal, endocrine, metabolic, acid-base and immune systems. In addition, circadian rhythms, including regular physical activity, directly influence the adaptive response of the microbiota. In conclusion, regular stimuli of moderate- and high-intensity physical activity promote greater diversity, abundance, resilience and versatility of the gut microbiota. This effect is highly beneficial for human health when healthy lifestyle habits including nutrition, hydration, rest, chronoregulation and physical activity.
Collapse
Affiliation(s)
- Jesús Álvarez-Herms
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain; Phymo® Lab, Physiology and Molecular Laboratory, Collado Hermoso, Segovia, Spain.
| | - Adrián Odriozola
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain
| |
Collapse
|
28
|
Qian JN, Kang YL, He YC, Hu HY. Topic Modeling Analysis of Chinese Medicine Literature on Gastroesophageal Reflux Disease: Insights into Potential Treatment. Chin J Integr Med 2024:10.1007/s11655-024-3800-y. [PMID: 38850480 DOI: 10.1007/s11655-024-3800-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2023] [Indexed: 06/10/2024]
Abstract
OBJECTIVE To analyze Chinese medicine (CM) prescriptions for gastroesophageal reflux disease (GERD), we model topics on GERD-related classical CM literature, providing insights into the potential treatment. METHODS Clinical guidelines were used to identify symptom terms for GERD, and CM literature from the database "Imedbooks" was retrieved for related prescriptions and their corresponding sources, indications, and other information. BERTopic was applied to identify the main topics and visualize the data. RESULTS A total of 36,207 entries are queried and 1,938 valid entries were acquired after manually filtering. Eight topics were identified by BERTopic, including digestion function abate, stomach flu, respiratory-related symptoms, gastric dysfunction, regurgitation and gastrointestinal dysfunction in pediatric patients, vomiting, stroke and alcohol accumulation are associated with the risk of GERD, vomiting and its causes, regurgitation, epigastric pain, and symptoms of heartburn. CONCLUSIONS Topic modeling provides an unbiased analysis of classical CM literature on GERD in a time-efficient and scale-efficient manner. Based on this analysis, we present a range of treatment options for relieving symptoms, including herbal remedies and non-pharmacological interventions such as acupuncture and dietary therapy.
Collapse
Affiliation(s)
- Jia-Nan Qian
- Department of Gastroenterology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Yan-Lan Kang
- Institute of AI and Robotics, Academy for Engineering & Technology, Fudan University, Shanghai, 200433, China
| | - You-Cheng He
- Clinical Research Center, Longhua Hospital, Shanghai, 200032, China
| | - Hong-Yi Hu
- Department of Gastroenterology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
29
|
Han A, Hudson-Paz C, Robinson BG, Becker L, Jacobson A, Kaltschmidt JA, Garrison JL, Bhatt AS, Monack DM. Temperature-dependent differences in mouse gut motility are mediated by stress. Lab Anim (NY) 2024; 53:148-159. [PMID: 38806681 PMCID: PMC11147774 DOI: 10.1038/s41684-024-01376-5] [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: 10/23/2023] [Accepted: 04/19/2024] [Indexed: 05/30/2024]
Abstract
Researchers have advocated elevating mouse housing temperatures from the conventional ~22 °C to the mouse thermoneutral point of 30 °C to enhance translational research. However, the impact of environmental temperature on mouse gastrointestinal physiology remains largely unexplored. Here we show that mice raised at 22 °C exhibit whole gut transit speed nearly twice as fast as those raised at 30 °C, primarily driven by a threefold increase in colon transit speed. Furthermore, gut microbiota composition differs between the two temperatures but does not dictate temperature-dependent differences in gut motility. Notably, increased stress signals from the hypothalamic-pituitary-adrenal axis at 22 °C have a pivotal role in mediating temperature-dependent differences in gut motility. Pharmacological and genetic depletion of the stress hormone corticotropin-releasing hormone slows gut motility in stressed 22 °C mice but has no comparable effect in relatively unstressed 30 °C mice. In conclusion, our findings highlight that colder mouse facility temperatures significantly increase gut motility through hormonal stress pathways.
Collapse
Affiliation(s)
- Alvin Han
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | | | - Beatriz G Robinson
- Neurosciences IDP Graduate Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Laren Becker
- Department of Medicine (Gastroenterology and Hepatology), Stanford University, Stanford, CA, USA
| | - Amanda Jacobson
- Genentech Inc., Research and Early Development, Immunology Discovery, South San Francisco, CA, USA
| | - Julia A Kaltschmidt
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Jennifer L Garrison
- Buck Institute for Research on Aging, Novato, CA, USA
- Global Consortium for Reproductive Longevity & Equality, Novato, CA, USA
| | - Ami S Bhatt
- Department of Medicine (Hematology, Blood and Marrow Transplantation), Stanford University, Stanford, CA, USA.
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
| | - Denise M Monack
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
30
|
Wang R, Bai B, Huang Y, Degen A, Mi J, Xue Y, Hao L. Yaks Are Dependent on Gut Microbiota for Survival in the Environment of the Qinghai Tibet Plateau. Microorganisms 2024; 12:1122. [PMID: 38930503 PMCID: PMC11205922 DOI: 10.3390/microorganisms12061122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
The yak (Poephagus grunniens) has evolved unique adaptations to survive the harsh environment of the Qinghai-Tibetan Plateau, while their gut microorganisms play a crucial role in maintaining the health of the animal. Gut microbes spread through the animal population not only by horizontal transmission but also vertically, which enhances microbial stability and inheritance between generations of the population. Homogenization of gut microbes in different animal species occurs in the same habitat, promoting interspecies coexistence. Using the yak as a model animal, this paper discusses the adaptive strategies under extreme environments, and how the gut microbes of the yak circulate throughout the Tibetan Plateau system, which not only affects other plateau animals such as plateau pikas, but can also have a profound impact on the health of people. By examining the relationships between yaks and their gut microbiota, this review offers new insights into the adaptation of yaks and their ecological niche on the Qinghai-Tibetan plateau.
Collapse
Affiliation(s)
- Runze Wang
- Key Laboratory of Plateau Grazing Animal Nutrition and Feed Science of Qinghai Province, State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining 810016, China; (R.W.); (B.B.)
| | - Binqiang Bai
- Key Laboratory of Plateau Grazing Animal Nutrition and Feed Science of Qinghai Province, State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining 810016, China; (R.W.); (B.B.)
| | - Yayu Huang
- PEGASE, INRAE, Institut Agro, 35590 Saint-Gilles, France;
| | - Allan Degen
- Desert Animal Adaptations and Husbandry, Wyler Department of Dryland Agriculture, Blaustein Institutes for Desert Research, Ben-Gurion University of the Negev, Beer Sheva 8410500, Israel;
| | - Jiandui Mi
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou 730000, China;
| | - Yanfeng Xue
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China;
| | - Lizhuang Hao
- Key Laboratory of Plateau Grazing Animal Nutrition and Feed Science of Qinghai Province, State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining 810016, China; (R.W.); (B.B.)
| |
Collapse
|
31
|
Wang M, Guo W, Chen JF. Caffeine: a potential mechanism for anti-obesity. Purinergic Signal 2024:10.1007/s11302-024-10022-1. [PMID: 38802651 DOI: 10.1007/s11302-024-10022-1] [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: 03/05/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024] Open
Abstract
Obesity refers to the excessive accumulation of fat caused by a long-term imbalance between energy intake (EI) and energy expenditure (EE). Over recent years, obesity has become a major public health challenge. Caffeine is a natural product that has been demonstrated to exert anti-obesity effects; however, the mechanisms responsible for the effect of caffeine on weight loss have yet to be fully elucidated. Most obesity-related deaths are due to cardiovascular disease. Recent research has demonstrated that caffeine can reduce the risk of death from cardiovascular disease; thus, it can be hypothesized that caffeine may represent a new therapeutic agent for weight loss. In this review, we synthesize data arising from clinical and animal studies over the last decade and discuss the potential mechanisms by which caffeine may induce weight loss, focusing particularly on increasing energy consumption, suppressing appetite, altering lipid metabolism, and influencing the gut microbiota. Finally, we summarize the major challenges associated with caffeine and anti-obesity research and highlight possible directions for future research and development.
Collapse
Affiliation(s)
- Meng Wang
- International Joint Research Center on Purinergic Signaling, School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Wei Guo
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Jiang-Fan Chen
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
32
|
Guo J, Zhang H, Hu H, Zhao T, Ji H, Ma L, Lu J, Yuan J, Xu B. Silent information regulator 2 deficiency exacerbates chronic cold exposure-induced colonic injury and p65 activation in mice. Gene 2024; 907:148276. [PMID: 38360128 DOI: 10.1016/j.gene.2024.148276] [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: 11/21/2023] [Revised: 01/18/2024] [Accepted: 02/08/2024] [Indexed: 02/17/2024]
Abstract
Cold is a common stressor that threatens colonic health by affecting internal homeostasis. From the literature, Silent information regulator 2 (SIRT2) may have important roles during cold stress, but this conjecture requires investigation. To address this knowledge gap, we investigated the effects of SIRT2 on colonic injury in chronically cold-exposure mice. In a previous study, we showed that SIRT2 regulated p65 activation after cold exposure. In the current study, mice were exposed to 4 °C for 3 h/day for 3 weeks to simulate a chronic cold exposure environment. Chronic cold exposure shortened colon length, disrupted tight junctions in colonic epithelial tissue, and disordered colonic flora. Chronic cold exposure also increased p65 acetylation levels, promoted nuclear factor (NF)-κB activation, and increased the expression of its downstream pro-inflammatory factors, while SIRT2 knockdown aggravated the consequences of tissue structure disruption and increased inflammatory factors brought about by chronic cold exposure to some extent, but could alleviate the downregulation of colonic tight junction-related proteins to some extent. We also observed direct SIRT2 regulatory effects toward p65, and in Caco-2 cells treated with lipopolysaccharide (LPS), SIRT2 knockdown increased p65 acetylation levels and pro-inflammatory factor expression, while SIRT2 overexpression reversed these phenomena. Therefore, SIRT2 deletion exacerbated chronic cold exposure-induced colonic injury and p65 activation in mice. Mechanistically, p65 modification by SIRT2 via deacetylation may affect NF-κB signaling. These findings suggest that SIRT2 is a key target of colonic health maintenance under chronic cold exposure conditions.
Collapse
Affiliation(s)
- Jingru Guo
- National Experimental Teaching Demonstration Center of Animal Medicine Foundation, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Huaixiu Zhang
- National Experimental Teaching Demonstration Center of Animal Medicine Foundation, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Huijie Hu
- National Experimental Teaching Demonstration Center of Animal Medicine Foundation, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Tianrui Zhao
- National Experimental Teaching Demonstration Center of Animal Medicine Foundation, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Hong Ji
- National Experimental Teaching Demonstration Center of Animal Medicine Foundation, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Li Ma
- National Experimental Teaching Demonstration Center of Animal Medicine Foundation, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Jingjing Lu
- National Experimental Teaching Demonstration Center of Animal Medicine Foundation, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Jianbin Yuan
- National Experimental Teaching Demonstration Center of Animal Medicine Foundation, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Bin Xu
- National Experimental Teaching Demonstration Center of Animal Medicine Foundation, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China.
| |
Collapse
|
33
|
Hassan NE, El-Masry SA, El Shebini SM, Ahmed NH, Mehanna NS, Abdel Wahed MM, Amine D, Hashish A, Selim M, Afify MAS, Alian K. Effect of weight loss program using prebiotics and probiotics on body composition, physique, and metabolic products: longitudinal intervention study. Sci Rep 2024; 14:10960. [PMID: 38744950 PMCID: PMC11094057 DOI: 10.1038/s41598-024-61130-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 05/02/2024] [Indexed: 05/16/2024] Open
Abstract
The relationship between gut microbiota and obesity has recently been an important subject for research as the gut microbiota is thought to affect body homeostasis including body weight and composition, intervening with pro and prebiotics is an intelligent possible way for obesity management. To evaluate the effect of hypo caloric adequate fiber regimen with probiotic supplementation and physical exercise, whether it will have a good impact on health, body composition, and physique among obese Egyptian women or has no significant effect. The enrolled 58 women, in this longitudinal follow-up intervention study; followed a weight loss eating regimen (prebiotic), including a low-carbohydrate adequate-fiber adequate-protein dietary pattern with decreased energy intake. They additionally received daily probiotic supplements in the form of yogurt and were instructed to exercise regularly for 3 months. Anthropometric measurements, body composition, laboratory investigations, and microbiota analysis were obtained before and after the 3 months weight loss program. Statistically highly significant differences in the anthropometry, body composition parameters: and obesity-related biomarkers (Leptin, ALT, and AST) between the pre and post-follow-up measurements at the end of the study as they were all decreased. The prebiotic and probiotic supplementation induced statistically highly significant alterations in the composition of the gut microbiota with increased relative abundance of Lactobacillus, Bifidobacteria, and Bacteroidetes and decreased relative abundance of Firmicutes and Firmicutes/Bacteroidetes Ratio. Hypo caloric adequate fiber regimen diet with probiotics positively impacts body composition and is effective for weight loss normalizing serum Leptin and AST.
Collapse
Affiliation(s)
- Nayera E Hassan
- Biological Anthropology Department, Medical Research and Clinical Studies Institute, National Research Centre, 33 El-Buhouth St., Dokki, Giza, 12622, Egypt
| | - Sahar A El-Masry
- Biological Anthropology Department, Medical Research and Clinical Studies Institute, National Research Centre, 33 El-Buhouth St., Dokki, Giza, 12622, Egypt.
| | - Salwa M El Shebini
- Nutrition and Food Science Department, Nutrition and Food Science Institute, National Research Centre, Giza, Egypt
| | - Nihad H Ahmed
- Nutrition and Food Science Department, Nutrition and Food Science Institute, National Research Centre, Giza, Egypt
| | - Nayra Sh Mehanna
- Dairy Science Department, Nutrition and Food Science Institute, National Research Centre, Giza, Egypt
| | - Mai Magdy Abdel Wahed
- Clinical and Chemical Pathology Department, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Darine Amine
- Biological Anthropology Department, Medical Research and Clinical Studies Institute, National Research Centre, 33 El-Buhouth St., Dokki, Giza, 12622, Egypt
| | - Adel Hashish
- Children with Special Needs Department, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Mohamed Selim
- Researches and Applications of Complementary Medicine Department, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Mahmoud A S Afify
- Biological Anthropology Department, Medical Research and Clinical Studies Institute, National Research Centre, 33 El-Buhouth St., Dokki, Giza, 12622, Egypt
| | - Khadija Alian
- Biological Anthropology Department, Medical Research and Clinical Studies Institute, National Research Centre, 33 El-Buhouth St., Dokki, Giza, 12622, Egypt
| |
Collapse
|
34
|
Kang M, Kang M, Yoo J, Lee J, Lee S, Yun B, Song M, Kim JM, Kim HW, Yang J, Kim Y, Oh S. Dietary supplementation with Lacticaseibacillus rhamnosus IDCC3201 alleviates sarcopenia by modulating the gut microbiota and metabolites in dexamethasone-induced models. Food Funct 2024; 15:4936-4953. [PMID: 38602003 DOI: 10.1039/d3fo05420a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
Probiotics can exert direct or indirect influences on various aspects of health claims by altering the composition of the gut microbiome and producing bioactive metabolites. The aim of this study was to examine the effect of Lacticaseibacillus rhamnosus IDCC3201 on skeletal muscle atrophy in dexamethasone-induced C2C12 cells and a mouse animal model. Dexamethasone treatment significantly reduced C2C12 muscle cell viability, myotube diameter, and levels of muscle atrophic markers (Atrogin-1 and MuRF-1). These effects were alleviated by conditioned media (CM) and cell extract (EX) derived from L. rhamnosus IDCC3201. In addition, we assessed the in vivo therapeutic effect of L. rhamnosus IDCC3201 in a mouse model of dexamethasone (DEX)-induced muscle atrophy. Supplementation with IDCC3201 resulted in significant enhancements in body composition, particularly in lean mass, muscle strength, and myofibril size, in DEX-induced muscle atrophy mice. In comparison to the DEX-treatment group, the normal and DEX + L. rhamnosus IDCC3201 groups showed a higher transcriptional level of myosin heavy chain family genes (MHC1, MHC1b, MHC2A, 2bB, and 2X) and a reduction in atrophic muscle makers. These analyses revealed that L. rhamnosus IDCC3201 supplementation led to increased production of branched-chain amino acids (BCAAs) and improved the Allobaculum genus within the gut microbiota of muscle atrophy-induced groups. Taken together, our findings suggest that L. rhamnosus IDCC3201 represents a promising dietary supplement with the potential to alleviate sarcopenia by modulating the gut microbiome and metabolites.
Collapse
Affiliation(s)
- Minkyoung Kang
- Department of Food and Nutrition, Jeonju University, Jeonju 55069, Republic of Korea
| | - Minji Kang
- Department of Food and Nutrition, Jeonju University, Jeonju 55069, Republic of Korea
| | - Jiseon Yoo
- Department of Food and Nutrition, Jeonju University, Jeonju 55069, Republic of Korea
| | - Juyeon Lee
- Department of Food and Nutrition, Jeonju University, Jeonju 55069, Republic of Korea
| | - Sujeong Lee
- Department of Food and Nutrition, Jeonju University, Jeonju 55069, Republic of Korea
| | - Bohyun Yun
- Honam National Institute of Biological Resources, Mokpo 58762, Republic of Korea
| | - Minho Song
- Department of Animal Science and Biotechnology, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Jun-Mo Kim
- Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Gyeonggi-do, Republic of Korea
| | - Hyung Wook Kim
- College of Life Sciences, Sejong University, Seoul 05006, Republic of Korea
| | - Jungwoo Yang
- Department of Microbiology, College of Medicine, Dongguk University, Gyeongju, 38066, Republic of Korea
| | - Younghoon Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Sangnam Oh
- Department of Food and Nutrition, Jeonju University, Jeonju 55069, Republic of Korea
| |
Collapse
|
35
|
Zhou H, Yan J, Zhou K, Ji P, Wei Y, Hua Y. Effects of Huangqi Gancao Decoction on intestinal immunity and microbiota in immunocompromised mice models. Front Pharmacol 2024; 15:1390170. [PMID: 38756377 PMCID: PMC11097664 DOI: 10.3389/fphar.2024.1390170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 04/04/2024] [Indexed: 05/18/2024] Open
Abstract
Background The classical medicinal formula Huangqi Gancao Decoction (HQGCD), originating from the medical book" Yi Lin Gai Cuo". Up to now, the studies focusing on the immunoenhancement effects of HQGCD are few, and the actionpathway is not yet clear. Method In this study, SPF male KM mice were utilized as a model for immunosuppression. Comprehensive observations were made regarding the general behavior and condition of the mice, in addition to monitoring fluctuations in body weight and food intake. The blood routine index was measured, and morphological changes in the ileum and colon tissues were examined. The level of secretory immunoglobulin A (sIgA), superoxide dismutase (SOD), and malondialdehyde (MDA) in ileum and colon tissues were quantified. Additionally, the bone marrow total DNA index was assessed. Flow cytometry analyzed the proportions of CD3⁺, CD4⁺, CD8⁺, and CD4+CD8+ double-positive (DP) T lymphocytes in small intestinal intraepithelial lymphocytes (IELs). Lastly, the composition and diversity of the cecal microbiota were evaluated using 16S rDNA sequencing technology. Results After HQGCD intervention, there were no significant changes in the mice's feed intake and body weight. However, the tissue structures of the ileum and colon showed recovery. In the blood routine index, there was an increase in the total white blood cell count, lymphocyte count, red blood cell count, hematocrit, and hemoglobin content. Additionally, the bone marrow total DNA index was elevated. Level of SOD and sIgA in ileum and colon tissues increased, while the level of MDA decreased. The proportions of CD3⁺ and CD4⁺ T lymphocytes within IELs increased, along with an increase in DP T lymphocytes in IELs (DP IELs), whereas the proportion of CD8⁺ T lymphocytes decreased. The cecal microbiota underwent changes, with an increase in the variety and number of beneficial microbiota. Conclusion HQGCD could restore the intestinal immune function of immunocompromised mice, and had a certain positive effect on cecal microbiota.
Collapse
Affiliation(s)
- Hai Zhou
- Tranditional Chinese Veterinary Medicine Laboratory, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Jianpeng Yan
- Tranditional Chinese Veterinary Medicine Laboratory, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
- Lanzhou Animal Disease Control Center, Lanzhou, Gansu, China
| | - Ke Zhou
- Tranditional Chinese Veterinary Medicine Laboratory, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Peng Ji
- Tranditional Chinese Veterinary Medicine Laboratory, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Yanming Wei
- Tranditional Chinese Veterinary Medicine Laboratory, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Yongli Hua
- Tranditional Chinese Veterinary Medicine Laboratory, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
| |
Collapse
|
36
|
Lv H, Xia S, He Y, Qiao C, Liu J, Guo J, Li S. Effect of chronic cold stress on gut microbial diversity, intestinal inflammation and pyroptosis in mice. J Physiol Biochem 2024; 80:465-477. [PMID: 38526704 DOI: 10.1007/s13105-024-01019-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 03/16/2024] [Indexed: 03/27/2024]
Abstract
Hypothermia is an essential environmental factor in gastrointestinal diseases, but the main molecular mechanisms of pathogenesis remain unclear. The current study sought to better understand how chronic cold stress affects gut damage and its underlying mechanisms. In this work, to establish chronic cold stress (CS)-induced intestinal injury model, mice were subjected to continuous cold exposure (4 °C) for 3 h per day for 3 weeks. Our results indicated that CS led to gut injury via inducing changes of heat shock proteins 70 (HSP70) and apoptosis-related (caspases-3, Bax and Bcl-2) proteins; enhancing expression of intestinal tight-related (ZO-1 and occludin) proteins; promoting releases of inducible nitric oxide synthase (iNOS), tumor necrosis factor-α (TNF-α), cyclooxygenase-2 (COX-2), high mobility group box 1 (HMGB1), interleukin1β (IL-1β), IL-18 and IL-6 inflammatory mediators in the ileum; and altering gut microbial diversity. Furthermore, persistent cold exposure resulted in the cleavage of pyroptosis-related Gasdermin D (GSDMD) protein by regulating the NLRP3/ASC/caspase-1 and caspase-11 pathway, and activation of toll-like receptor 4 (TLR4)/myeloid differentiation factor 88 (MyD88)-mediated nuclear factor kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways, which are strongly associated with changes in gut microbiota diversity. Taken together, these investigations provide new insights into the increased risk of intestinal disorders at extremely low temperatures and establish a theoretical foundation for the advancement of novel pharmaceutical interventions targeting cold-related ailments.
Collapse
Affiliation(s)
- Hongming Lv
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs; Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases; College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Shijie Xia
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs; Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases; College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Yuxi He
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs; Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases; College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Chunyu Qiao
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs; Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases; College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Jiahe Liu
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs; Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases; College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Jingru Guo
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs; Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases; College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, China.
| | - Shize Li
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs; Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases; College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, China.
| |
Collapse
|
37
|
Young AP, Denovan-Wright EM. JAK1/2 Regulates Synergy Between Interferon Gamma and Lipopolysaccharides in Microglia. J Neuroimmune Pharmacol 2024; 19:14. [PMID: 38642237 DOI: 10.1007/s11481-024-10115-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 04/01/2024] [Indexed: 04/22/2024]
Abstract
Microglia, the resident immune cells of the brain, regulate neuroinflammation which can lead to secondary neuronal damage and cognitive impairment under pathological conditions. Two of the many molecules that can elicit an inflammatory response from microglia are lipopolysaccharide (LPS), a component of gram-negative bacteria, and interferon gamma (IFNγ), an endogenous pro-inflammatory cytokine. We thoroughly examined the concentration-dependent relationship between LPS from multiple bacterial species and IFNγ in cultured microglia and macrophages. We measured the effects that these immunostimulatory molecules have on pro-inflammatory activity of microglia and used a battery of signaling inhibitors to identify the pathways that contribute to the microglial response. We found that LPS and IFNγ interacted synergistically to induce a pro-inflammatory phenotype in microglia, and that inhibition of JAK1/2 completely blunted the response. We determined that this synergistic action of LPS and IFNγ was likely dependent on JNK and Akt signaling rather than typical pro-inflammatory mediators such as NF-κB. Finally, we demonstrated that LPS derived from Escherichia coli, Klebsiella pneumoniae, and Akkermansia muciniphila can elicit different inflammatory responses from microglia and macrophages, but these responses could be consistently prevented using ruxolitinib, a JAK1/2 inhibitor. Collectively, this work reveals a mechanism by which microglia may become hyperactivated in response to the combination of LPS and IFNγ. Given that elevations in circulating LPS and IFNγ occur in a wide variety of pathological conditions, it is critical to understand the pharmacological interactions between these molecules to develop safe and effective treatments to suppress this process.
Collapse
Affiliation(s)
- Alexander P Young
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada.
| | | |
Collapse
|
38
|
Wang Z, Wu Y, Li X, Ji X, Liu W. The gut microbiota facilitate their host tolerance to extreme temperatures. BMC Microbiol 2024; 24:131. [PMID: 38643098 PMCID: PMC11031955 DOI: 10.1186/s12866-024-03277-6] [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: 12/11/2023] [Accepted: 03/25/2024] [Indexed: 04/22/2024] Open
Abstract
BACKGROUND Exposure to extreme cold or heat temperature is one leading cause of weather-associated mortality and morbidity in animals. Emerging studies demonstrate that the microbiota residing in guts act as an integral factor required to modulate host tolerance to cold or heat exposure, but common and unique patterns of animal-temperature associations between cold and heat have not been simultaneously examined. Therefore, we attempted to investigate the roles of gut microbiota in modulating tolerance to cold or heat exposure in mice. RESULTS The results showed that both cold and heat acutely change the body temperature of mice, but mice efficiently maintain their body temperature at conditions of chronic extreme temperatures. Mice adapt to extreme temperatures by adjusting body weight gain, food intake and energy harvest. Fascinatingly, 16 S rRNA sequencing shows that extreme temperatures result in a differential shift in the gut microbiota. Moreover, transplantation of the extreme-temperature microbiota is sufficient to enhance host tolerance to cold and heat, respectively. Metagenomic sequencing shows that the microbiota assists their hosts in resisting extreme temperatures through regulating the host insulin pathway. CONCLUSIONS Our findings highlight that the microbiota is a key factor orchestrating the overall energy homeostasis under extreme temperatures, providing an insight into the interaction and coevolution of hosts and gut microbiota.
Collapse
Affiliation(s)
- Ziguang Wang
- School of Plant Protection, Anhui Province Key Laboratory of Crop Integrated Pest Management, Anhui Province Engineering Laboratory for Green Pesticide Development and Application, Anhui Agricultural University, Hefei, China
- First Clinical Medical College, Mudanjiang Medical College, Mudanjiang, China
| | - Yujie Wu
- School of Plant Protection, Anhui Province Key Laboratory of Crop Integrated Pest Management, Anhui Province Engineering Laboratory for Green Pesticide Development and Application, Anhui Agricultural University, Hefei, China
| | - Xinxin Li
- China-Japan Union Hospital, Jilin University, Changchun, China
| | - Xiaowen Ji
- School of Plant Protection, Anhui Province Key Laboratory of Crop Integrated Pest Management, Anhui Province Engineering Laboratory for Green Pesticide Development and Application, Anhui Agricultural University, Hefei, China.
| | - Wei Liu
- School of Plant Protection, Anhui Province Key Laboratory of Crop Integrated Pest Management, Anhui Province Engineering Laboratory for Green Pesticide Development and Application, Anhui Agricultural University, Hefei, China.
| |
Collapse
|
39
|
Chen S, Hu Z, Tang J, Zhu H, Zheng Y, Xiao J, Xu Y, Wang Y, Luo Y, Mo X, Wu Y, Guo J, Zhang Y, Luo H. High temperature and humidity in the environment disrupt bile acid metabolism, the gut microbiome, and GLP-1 secretion in mice. Commun Biol 2024; 7:465. [PMID: 38632312 PMCID: PMC11024098 DOI: 10.1038/s42003-024-06158-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: 06/23/2023] [Accepted: 04/05/2024] [Indexed: 04/19/2024] Open
Abstract
High temperature and humidity in the environment are known to be associated with discomfort and disease, yet the underlying mechanisms remain unclear. We observed a decrease in plasma glucagon-like peptide-1 levels in response to high-temperature and humidity conditions. Through 16S rRNA gene sequencing, alterations in the gut microbiota composition were identified following exposure to high temperature and humidity conditions. Notably, changes in the gut microbiota have been implicated in bile acid synthesis. Further analysis revealed a decrease in lithocholic acid levels in high-temperature and humidity conditions. Subsequent in vitro experiments demonstrated that lithocholic acid increases glucagon-like peptide-1 secretion in NCI-H716 cells. Proteomic analysis indicated upregulation of farnesoid X receptor expression in the ileum. In vitro experiments revealed that the combination of lithocholic acid with farnesoid X receptor inhibitors resulted in a significant increase in GLP-1 levels compared to lithocholic acid alone. In this study, we elucidate the mechanism by which reduced lithocholic acid suppresses glucagon-like peptide 1 via farnesoid X receptor activation under high-temperature and humidity condition.
Collapse
Affiliation(s)
- Song Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zongren Hu
- Department of Rehabilitation and Healthcare, Hunan University of Medicine, Huaihua, China
| | - Jianbang Tang
- Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Chinese Medicine, Zhongshan, China
| | | | - Yuhua Zheng
- School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiedong Xiao
- School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Youhua Xu
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, Macao, China
| | - Yao Wang
- School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yi Luo
- School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoying Mo
- School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yalan Wu
- School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianwen Guo
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China.
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Yongliang Zhang
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Immunology Programme, The Life Science Institute, National University of Singapore, Singapore, Singapore.
| | - Huanhuan Luo
- School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China.
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
40
|
Zhu XM, Chen JQ, Du Y, Lin CX, Qu YF, Lin LH, Ji X. Microbial communities are thermally more sensitive in warm-climate lizards compared with their cold-climate counterparts. Front Microbiol 2024; 15:1374209. [PMID: 38686106 PMCID: PMC11056556 DOI: 10.3389/fmicb.2024.1374209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 04/01/2024] [Indexed: 05/02/2024] Open
Abstract
Environmental temperature affects the composition, structure, and function of the gut microbial communities in host animals. To elucidate the role of gut microbiota in thermal adaptation, we designed a 2 species × 3 temperatures experiment, whereby we acclimated adult males of two agamid lizard species (warm-climate Leiolepis reevesii and cold-climate Phrynocephalus przewalskii) to 20, 28, and 36°C for 2 weeks and then collected their fecal and small-intestinal samples to analyze and compare the microbiota using 16S rRNA gene amplicon sequencing technology. The fecal microbiota displayed more pronounced interspecific differences in microbial community than the small-intestinal microbiota in the two species occurring in thermally different regions. The response of fecal and small-intestinal microbiota to temperature increase or decrease differed between the two species, with more bacterial taxa affected by acclimation temperature in L. reevesii than in P. przewalskii. Both species, the warm-climate species in particular, could cope with temperature change by adjusting the relative abundance of functional categories associated with metabolism and environmental information processing. Functional genes associated with carbohydrate metabolism were enhanced in P. przewalskii, suggesting the contribution of the fecal microbiota to cold-climate adaptation in P. przewalskii. Taken together, our results validate the two hypotheses tested, of which one suggests that the gut microbiota should help lizards adapt to thermal environments in which they live, and the other suggests that microbial communities should be thermally more sensitive in warm-climate lizards than in cold-climate lizards.
Collapse
Affiliation(s)
- Xia-Ming Zhu
- College of Life Sciences, Nanjing Normal University, Nanjing, China
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Jun-Qiong Chen
- College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Yu Du
- Hainan Key Laboratory of Herpetological Research, College of Fisheries and Life Sciences, Hainan Tropical Ocean University, Sanya, China
| | - Chi-Xian Lin
- Hainan Key Laboratory of Herpetological Research, College of Fisheries and Life Sciences, Hainan Tropical Ocean University, Sanya, China
| | - Yan-Fu Qu
- College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Long-Hui Lin
- Herpetological Research Center, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Xiang Ji
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| |
Collapse
|
41
|
Xing PY, Agrawal R, Jayaraman A, Martin KA, Zhang GW, Ngu EL, Faylon LE, Kjelleberg S, Rice SA, Wang Y, Bello AT, Holmes E, Nicholson JK, Whiley L, Pettersson S. Microbial Indoles: Key Regulators of Organ Growth and Metabolic Function. Microorganisms 2024; 12:719. [PMID: 38674663 PMCID: PMC11052216 DOI: 10.3390/microorganisms12040719] [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: 03/07/2024] [Revised: 03/26/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
Gut microbes supporting body growth are known but the mechanisms are less well documented. Using the microbial tryptophan metabolite indole, known to regulate prokaryotic cell division and metabolic stress conditions, we mono-colonized germ-free (GF) mice with indole-producing wild-type Escherichia coli (E. coli) or tryptophanase-encoding tnaA knockout mutant indole-non-producing E. coli. Indole mutant E. coli mice showed multiorgan growth retardation and lower levels of glycogen, cholesterol, triglycerides, and glucose, resulting in an energy deficiency despite increased food intake. Detailed analysis revealed a malfunctioning intestine, enlarged cecum, and reduced numbers of enterochromaffin cells, correlating with a metabolic phenotype consisting of impaired gut motility, diminished digestion, and lower energy harvest. Furthermore, indole mutant mice displayed reduction in serum levels of tricarboxylic acid (TCA) cycle intermediates and lipids. In stark contrast, a massive increase in serum melatonin was observed-frequently associated with accelerated oxidative stress and mitochondrial dysfunction. This observational report discloses functional roles of microbe-derived indoles regulating multiple organ functions and extends our previous report of indole-linked regulation of adult neurogenesis. Since indoles decline by age, these results imply a correlation with age-linked organ decline and levels of indoles. Interestingly, increased levels of indole-3-acetic acid, a known indole metabolite, have been shown to correlate with younger biological age, further supporting a link between biological age and levels of microbe-derived indole metabolites. The results presented in this resource paper will be useful for the future design of food intervention studies to reduce accelerated age-linked organ decline.
Collapse
Affiliation(s)
- Peter Yuli Xing
- Singapore Centre for Environmental Life Sciences Engineering, Singapore 637551, Singapore
- Interdisciplinary Graduate School, Nanyang Technological University, Singapore 637335, Singapore
| | - Ruchi Agrawal
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Anusha Jayaraman
- ASEAN Microbiome Nutrition Centre, National Neuroscience Institute, Singapore 308433, Singapore
| | - Katherine Ann Martin
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - George Wei Zhang
- ASEAN Microbiome Nutrition Centre, National Neuroscience Institute, Singapore 308433, Singapore
| | - Ee Ling Ngu
- ASEAN Microbiome Nutrition Centre, National Neuroscience Institute, Singapore 308433, Singapore
- Faculty of Medical Sciences, Sunway University, Subang Jaya 47500, Selangor, Malaysia
| | - Llanto Elma Faylon
- Singapore Centre for Environmental Life Sciences Engineering, Singapore 637551, Singapore
| | - Staffan Kjelleberg
- Singapore Centre for Environmental Life Sciences Engineering, Singapore 637551, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - Scott A. Rice
- Singapore Centre for Environmental Life Sciences Engineering, Singapore 637551, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - Yulan Wang
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
- Singapore Phenome Centre, Singapore 636921, Singapore
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London SW7 2AZ, UK
| | - Adesola T. Bello
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London SW7 2AZ, UK
- UK Dementia Research Institute, Imperial College London, London W1T 7NF, UK
| | - Elaine Holmes
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London SW7 2AZ, UK
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Perth, WA 6150, Australia
| | - Jeremy K. Nicholson
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Perth, WA 6150, Australia
- Institute of Global Health Innovation, Imperial College London, London SW7 2NA, UK
| | - Luke Whiley
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Perth, WA 6150, Australia
- Perron Institute, Nedlands, WA 6009, Australia
| | - Sven Pettersson
- ASEAN Microbiome Nutrition Centre, National Neuroscience Institute, Singapore 308433, Singapore
- Faculty of Medical Sciences, Sunway University, Subang Jaya 47500, Selangor, Malaysia
- Karolinska Institutet, 171 77 Solna, Sweden
- Department of Microbiology and Immunology, National University Singapore, Singapore 117545, Singapore
| |
Collapse
|
42
|
Ma C, Huang Z, Feng X, Memon FU, Cui Y, Duan X, Zhu J, Tettamanti G, Hu W, Tian L. Selective breeding of cold-tolerant black soldier fly (Hermetia illucens) larvae: Gut microbial shifts and transcriptional patterns. WASTE MANAGEMENT (NEW YORK, N.Y.) 2024; 177:252-265. [PMID: 38354633 DOI: 10.1016/j.wasman.2024.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 12/29/2023] [Accepted: 02/05/2024] [Indexed: 02/16/2024]
Abstract
The larvae of black soldier fly (BSFL) convert organic waste into insect proteins used as feedstuff for livestock and aquaculture. BSFL production performance is considerably reduced during winter season. Herein, the intraspecific diversity of ten commercial BSF colonies collected in China was evaluated. The Bioforte colony was subjected to selective breeding at 12 °C and 16 °C to develop cold-tolerant BSF with improved production performance. After breeding for nine generations, the weight of larvae, survival rate, and the dry matter conversion rate significantly increased. Subsequently, intestinal microbiota in the cold-tolerant strain showed that bacteria belonging to Morganella, Dysgonomonas, Salmonella, Pseudochrobactrum, and Klebsiella genera were highly represented in the 12 °C bred, while those of Acinetobacter, Pseudochrobactrum, Enterococcus, Comamonas, and Leucobacter genera were significantly represented in the 16 °C bred group. Metagenomic revealed that several animal probiotics of the Enterococcus and Vagococcus genera were greatly enriched in the gut of larvae bred at 16 °C. Moreover, bacterial metabolic pathways including carbohydrate, lipid, amino acids, and cofactors and vitamins, were significantly increased, while organismal systems and human diseases was decreased in the 16 °C bred group. Transcriptomic analysis revealed that the upregulated differentially expressed genes in the 16 °C bred groups mainly participated in Autophagy-animal, AMPK signaling pathway, mTOR signaling pathway, Wnt signaling pathway, FoxO signaling pathway, Hippo signaling pathway at day 34 under 16 °C conditions, suggesting their significant role in the survival of BSFL. Taken together, these results shed lights on the role of intestinal microflora and gene pathways in the adaptation of BSF larvae to cold stress.
Collapse
Affiliation(s)
- Chong Ma
- Guangdong Provincial Key Lab of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Bioforte Biotechnology (Shenzhen) Co., Ltd., Shenzhen 518118, China
| | - Zhijun Huang
- Guangdong Provincial Key Lab of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Bioforte Biotechnology (Shenzhen) Co., Ltd., Shenzhen 518118, China
| | - Xingbao Feng
- Guangdong Provincial Key Lab of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Bioforte Biotechnology (Shenzhen) Co., Ltd., Shenzhen 518118, China
| | - Fareed Uddin Memon
- Guangdong Provincial Key Lab of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Bioforte Biotechnology (Shenzhen) Co., Ltd., Shenzhen 518118, China
| | - Ying Cui
- Guangdong Provincial Key Lab of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Bioforte Biotechnology (Shenzhen) Co., Ltd., Shenzhen 518118, China
| | - Xinyu Duan
- Guangdong Provincial Key Lab of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Bioforte Biotechnology (Shenzhen) Co., Ltd., Shenzhen 518118, China
| | - Jianfeng Zhu
- Guangdong Provincial Key Lab of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Bioforte Biotechnology (Shenzhen) Co., Ltd., Shenzhen 518118, China
| | - Gianluca Tettamanti
- Department of Biotechnology and Life Sciences, University of Insubria, Varese 21100, Italy; Interuniversity Center for Studies on Bioinspired Agro-environmental Technology (BAT Center), University of Napoli Federico II, 80055 Portici, Italy
| | - Wenfeng Hu
- Guangdong Provincial Key Lab of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Bioforte Biotechnology (Shenzhen) Co., Ltd., Shenzhen 518118, China; Laboratory of Applied Microbiology, College of Food Science, South China Agricultural University, Guangdong 510642, China
| | - Ling Tian
- Guangdong Provincial Key Lab of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Bioforte Biotechnology (Shenzhen) Co., Ltd., Shenzhen 518118, China.
| |
Collapse
|
43
|
Mancin E, Maltecca C, Huang YJ, Mantovani R, Tiezzi F. A first characterization of the microbiota-resilience link in swine. MICROBIOME 2024; 12:53. [PMID: 38486255 PMCID: PMC10941389 DOI: 10.1186/s40168-024-01771-7] [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: 09/16/2023] [Accepted: 01/30/2024] [Indexed: 03/17/2024]
Abstract
BACKGROUND The gut microbiome plays a crucial role in understanding complex biological mechanisms, including host resilience to stressors. Investigating the microbiota-resilience link in animals and plants holds relevance in addressing challenges like adaptation of agricultural species to a warming environment. This study aims to characterize the microbiota-resilience connection in swine. As resilience is not directly observable, we estimated it using four distinct indicators based on daily feed consumption variability, assuming animals with greater intake variation may face challenges in maintaining stable physiological status. These indicators were analyzed both as linear and categorical variables. In our first set of analyses, we explored the microbiota-resilience link using PERMANOVA, α-diversity analysis, and discriminant analysis. Additionally, we quantified the ratio of estimated microbiota variance to total phenotypic variance (microbiability). Finally, we conducted a Partial Least Squares-Discriminant Analysis (PLS-DA) to assess the classification performance of the microbiota with indicators expressed in classes. RESULTS This study offers four key insights. Firstly, among all indicators, two effectively captured resilience. Secondly, our analyses revealed robust relationship between microbial composition and resilience in terms of both composition and richness. We found decreased α-diversity in less-resilient animals, while specific amplicon sequence variants (ASVs) and KEGG pathways associated with inflammatory responses were negatively linked to resilience. Thirdly, considering resilience indicators in classes, we observed significant differences in microbial composition primarily in animals with lower resilience. Lastly, our study indicates that gut microbial composition can serve as a reliable biomarker for distinguishing individuals with lower resilience. CONCLUSION Our comprehensive analyses have highlighted the host-microbiota and resilience connection, contributing valuable insights to the existing scientific knowledge. The practical implications of PLS-DA and microbiability results are noteworthy. PLS-DA suggests that host-microbiota interactions could be utilized as biomarkers for monitoring resilience. Furthermore, the microbiability findings show that leveraging host-microbiota insights may improve the identification of resilient animals, supporting their adaptive capacity in response to changing environmental conditions. These practical implications offer promising avenues for enhancing animal well-being and adaptation strategies in the context of environmental challenges faced by livestock populations. Video Abstract.
Collapse
Affiliation(s)
- Enrico Mancin
- Department of Agronomy, Animals and Environment, (DAFNAE), Food, Natural Resources, University of Padova, Viale del Università 14, 35020, Legnaro (Padova), Italy
| | - Christian Maltecca
- Department of Animal Science, North Carolina State University, Raleigh, NC, 27695, USA
- Department of Agriculture, Food, Environment and Forestry (DAGRI), University of Florence, Piazzale delle Cascine 18, 50144, Firenze, Italy
| | - Yi Jian Huang
- Smithfield Premium Genetics, Rose Hill, NC, 28458, USA
| | - Roberto Mantovani
- Department of Agronomy, Animals and Environment, (DAFNAE), Food, Natural Resources, University of Padova, Viale del Università 14, 35020, Legnaro (Padova), Italy
| | - Francesco Tiezzi
- Department of Agriculture, Food, Environment and Forestry (DAGRI), University of Florence, Piazzale delle Cascine 18, 50144, Firenze, Italy.
| |
Collapse
|
44
|
Zhou E, Zhang L, He L, Xiao Y, Zhang K, Luo B. Cold exposure, gut microbiota and health implications: A narrative review. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 916:170060. [PMID: 38242473 DOI: 10.1016/j.scitotenv.2024.170060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 01/07/2024] [Accepted: 01/08/2024] [Indexed: 01/21/2024]
Abstract
Temperature has been recognized as an important environmental factor affecting the composition and function of gut microbiota (GM). Although research on high-temperature impacts has been well studied, knowledge about the effect of cold exposure on GM remains limited. This narrative review aims to synthesize the latest scientific findings on the impact of cold exposure on mammalian GM, and its potential health implications. Chronic cold exposure could disrupt the α-diversity and the composition of GM in both experimental animals and wild-living hosts. Meanwhile, cold exposure could impact gut microbial metabolites, such as short-chain fatty acids. We also discussed plausible biological pathways and mechanisms by which cold-induced changes may impact host health, including metabolic homeostasis, fitness and thermogenesis, through the microbiota-gut-brain axis. Intriguingly, alterations in GM may provide a tool for favorably modulating the host response to the cold temperature. Finally, current challenges and future perspectives are discussed, emphasizing the need for translational research in humans. GM could be manipulated by utilizing nutritional strategies, such as probiotics and prebiotics, to deal with cold-related health issues and enhance well-being in populations living or working in cold environments.
Collapse
Affiliation(s)
- Erkai Zhou
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Ling Zhang
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Li He
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Ya Xiao
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Kai Zhang
- Department of Environmental Health Sciences, School of Public Health, University at Albany, State University of New York, Rensselaer, NY 12144, USA
| | - Bin Luo
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, China.
| |
Collapse
|
45
|
Chen PC, Tsai TP, Liao YC, Liao YC, Cheng HW, Weng YH, Lin CM, Kao CY, Tai CC, Ruan JW. Intestinal dual-specificity phosphatase 6 regulates the cold-induced gut microbiota remodeling to promote white adipose browning. NPJ Biofilms Microbiomes 2024; 10:22. [PMID: 38480743 PMCID: PMC10937957 DOI: 10.1038/s41522-024-00495-8] [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: 09/19/2023] [Accepted: 02/29/2024] [Indexed: 03/17/2024] Open
Abstract
Gut microbiota rearrangement induced by cold temperature is crucial for browning in murine white adipose tissue. This study provides evidence that DUSP6, a host factor, plays a critical role in regulating cold-induced gut microbiota rearrangement. When exposed to cold, the downregulation of intestinal DUSP6 increased the capacity of gut microbiota to produce ursodeoxycholic acid (UDCA). The DUSP6-UDCA axis is essential for driving Lachnospiraceae expansion in the cold microbiota. In mice experiencing cold-room temperature (CR) transitions, prolonged DUSP6 inhibition via the DUSP6 inhibitor (E/Z)-BCI maintained increased cecal UDCA levels and cold-like microbiota networks. By analyzing DUSP6-regulated microbiota dynamics in cold-exposed mice, we identified Marvinbryantia as a genus whose abundance increased in response to cold exposure. When inoculated with human-origin Marvinbryantia formatexigens, germ-free recipient mice exhibited significantly enhanced browning phenotypes in white adipose tissue. Moreover, M. formatexigens secreted the methylated amino acid Nε-methyl-L-lysine, an enriched cecal metabolite in Dusp6 knockout mice that reduces adiposity and ameliorates nonalcoholic steatohepatitis in mice. Our work revealed that host-microbiota coadaptation to cold environments is essential for regulating the browning-promoting gut microbiome.
Collapse
Affiliation(s)
- Pei-Chen Chen
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Tzu-Pei Tsai
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Yi-Chu Liao
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli, 35053, Taiwan
| | - Yu-Chieh Liao
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli, 35053, Taiwan
| | - Hung-Wei Cheng
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli, 35053, Taiwan
| | - Yi-Hsiu Weng
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Chiao-Mei Lin
- Immunology Research Center, National Health Research Institutes, Zhunan, Miaoli, 35053, Taiwan
| | - Cheng-Yuan Kao
- Immunology Research Center, National Health Research Institutes, Zhunan, Miaoli, 35053, Taiwan
| | | | - Jhen-Wei Ruan
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan.
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan.
- Research Center for Medical Laboratory Biotechnology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan.
| |
Collapse
|
46
|
Cani PD, Van Hul M. Gut microbiota in overweight and obesity: crosstalk with adipose tissue. Nat Rev Gastroenterol Hepatol 2024; 21:164-183. [PMID: 38066102 DOI: 10.1038/s41575-023-00867-z] [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] [Accepted: 10/27/2023] [Indexed: 03/02/2024]
Abstract
Overweight and obesity are characterized by excessive fat mass accumulation produced when energy intake exceeds energy expenditure. One plausible way to control energy expenditure is to modulate thermogenic pathways in white adipose tissue (WAT) and/or brown adipose tissue (BAT). Among the different environmental factors capable of influencing host metabolism and energy balance, the gut microbiota is now considered a key player. Following pioneering studies showing that mice lacking gut microbes (that is, germ-free mice) or depleted of their gut microbiota (that is, using antibiotics) developed less adipose tissue, numerous studies have investigated the complex interactions existing between gut bacteria, some of their membrane components (that is, lipopolysaccharides), and their metabolites (that is, short-chain fatty acids, endocannabinoids, bile acids, aryl hydrocarbon receptor ligands and tryptophan derivatives) as well as their contribution to the browning and/or beiging of WAT and changes in BAT activity. In this Review, we discuss the general physiology of both WAT and BAT. Subsequently, we introduce how gut bacteria and different microbiota-derived metabolites, their receptors and signalling pathways can regulate the development of adipose tissue and its metabolic capacities. Finally, we describe the key challenges in moving from bench to bedside by presenting specific key examples.
Collapse
Affiliation(s)
- Patrice D Cani
- Metabolism and Nutrition Research Group (MNUT), Louvain Drug Research Institute (LDRI), UCLouvain, Université catholique de Louvain, Brussels, Belgium.
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium.
- Institute of Experimental and Clinical Research (IREC), UCLouvain, Université catholique de Louvain, Brussels, Belgium.
| | - Matthias Van Hul
- Metabolism and Nutrition Research Group (MNUT), Louvain Drug Research Institute (LDRI), UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium
| |
Collapse
|
47
|
Wang H, Ülgen M, Trajkovski M. Importance of temperature on immuno-metabolic regulation and cancer progression. FEBS J 2024; 291:832-845. [PMID: 36152006 DOI: 10.1111/febs.16632] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/01/2022] [Accepted: 09/20/2022] [Indexed: 11/29/2022]
Abstract
Cancer immunotherapies emerge as promising strategies for restricting tumour growth. The tumour microenvironment (TME) has a major impact on the anti-tumour immune response and on the efficacy of the immunotherapies. Recent studies have linked changes in the ambient temperature with particular immuno-metabolic reprogramming and anti-cancer immune response in laboratory animals. Here, we describe the energetic balance of the organism during change in temperature, and link this to the immune alterations that could be of relevance for cancer, as well as for other human diseases. We highlight the contribution of the gut microbiota in modifying this interaction. We describe the overall metabolic response and underlying mechanisms of tumourigenesis in mouse models at varying ambient temperatures and shed light on their potential importance in developing therapeutics against cancer.
Collapse
Affiliation(s)
- Haiping Wang
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Centre Medical Universitaire (CMU), University of Geneva, Geneva, Switzerland
- Faculty of Medicine, Diabetes Center, University of Geneva, Geneva, Switzerland
| | - Melis Ülgen
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Centre Medical Universitaire (CMU), University of Geneva, Geneva, Switzerland
- Faculty of Medicine, Diabetes Center, University of Geneva, Geneva, Switzerland
| | - Mirko Trajkovski
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Centre Medical Universitaire (CMU), University of Geneva, Geneva, Switzerland
- Faculty of Medicine, Diabetes Center, University of Geneva, Geneva, Switzerland
| |
Collapse
|
48
|
Shuai He, Zhang KH, Jin QY, Wang QJ, Huang J, Li JJ, Guo Y, Liu P, Liu ZY, Liu D, Geng SX, Li Q, Li MY, Liu M, Wu ZH. The effects of ambient temperature and feeding regimens on cecum bacteria composition and circadian rhythm in growing rabbits. Front Microbiol 2024; 15:1344992. [PMID: 38476945 PMCID: PMC10927733 DOI: 10.3389/fmicb.2024.1344992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/05/2024] [Indexed: 03/14/2024] Open
Abstract
Seasonal environmental shifts and improper eating habits are the important causes of diarrhea in children and growing animals. Whether adjusting feeding time at varying temperatures can modify cecal bacterial structure and improve diarrhea remains unknown. Three batches growing rabbits with two groups per batch were raised under different feeding regimens (fed at daytime vs. nighttime) in spring, summer and winter separately, and contents were collected at six time points in 1 day and used 16S rRNA sequencing to investigate the effects of feeding regimens and season on the composition and circadian rhythms of cecum bacteria. Randomized forest regression screened 12 genera that were significantly associated with seasonal ambient temperature changes. Nighttime feeding reduced the abundance of the conditionally pathogenic bacteria Desulfovibrio and Alistipes in summer and Campylobacter in winter. And also increases the circadian rhythmic Amplicon Sequence Variants in the cecum, enhancing the rhythm of bacterial metabolic activity. This rhythmic metabolic profile of cecum bacteria may be conducive to the digestion and absorption of nutrients in the host cecum. In addition, this study has identified 9 genera that were affected by the combination of seasons and feeding time. In general, we found that seasons and feeding time and their combinations affect cecum composition and circadian rhythms, and that daytime feeding during summer and winter disrupts the balance of cecum bacteria of growing rabbits, which may adversely affect cecum health and induce diarrhea risk.
Collapse
Affiliation(s)
- Shuai He
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Ke-Hao Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Qiong-Yu Jin
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Qiang-Jun Wang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Jie Huang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jun-Jiao Li
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
- Handan Livestock Technology Extension Station, Handan, China
| | - Yao Guo
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Peng Liu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhong-Ying Liu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Dan Liu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shi-Xia Geng
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Qin Li
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Ming-Yong Li
- National Rabbit Industry Technology System Qingdao Comprehensive Experimental Station, Qingdao, China
| | - Man Liu
- National Rabbit Industry Technology System Qingdao Comprehensive Experimental Station, Qingdao, China
| | - Zhong-Hong Wu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
49
|
Luo T, Zhu J, Li K, Li Y, Li J, Chen Y, Shi H. Crosstalk between innate immunity and rumen-fecal microbiota under the cold stress in goats. Front Immunol 2024; 15:1363664. [PMID: 38476231 PMCID: PMC10928366 DOI: 10.3389/fimmu.2024.1363664] [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: 01/03/2024] [Accepted: 02/08/2024] [Indexed: 03/14/2024] Open
Abstract
The balance of the microbiome, which is sensitive to temperature changes, plays a crucial role in maintaining overall health and reducing the risk of diseases. However, the specific mechanisms by which immunity and microbiota interact to adapt to cold stress have yet to be addressed. In this study, Nanjiang Yellow goats were chosen as a model and sampled during the cold (winter, cold stress) and warm (spring) seasons, respectively. Analyses of serum immune factors, as well as the composition of rumen and fecal microbial communities, were conducted to explore the crosstalk between microbiota and innate immunity under cold stress. Significantly increased levels of IgA (P < 0.01) were observed in the cold season compared to the warm season. Conversely, the levels of IL-2 (P = 0.02) and IL-6 (P < 0.01) diminished under cold stress. However, no significant differences were observed in IgG (P = 0.89), IgM (P = 0.42), and IL-4 (P = 0.56). While there were no significant changes in the diversity of bacterial communities between the warm and cold seasons, positive correlations between serum IgA, IL-2, IL-6 concentrations and several genera were observed. Furthermore, the weighted gene co-expression network analysis indicated that the microbiota enriched in the MEbrown module positively correlated with IgA, while the microbiota enriched in the MEblue module positively correlated with IL-2 and IL-6. The strong correlation between certain probiotics, including Alistipes, Bacteroides, Blautia, and Prevotellaceae_UCG.004, and the concentration of IL-2, and IL-6 suggests their potential role in immunomodulatory properties. This study provides valuable insights into the crosstalk between microbial communities and immune responses under the challenge of cold stress. Further studies on the immunomodulatory properties of these probiotics would contribute to the development of strategies to enhance the stress resistance of animals for improved overall health and survival.
Collapse
Affiliation(s)
- Tao Luo
- Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Jiangjiang Zhu
- Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Chengdu, China
| | - Kerui Li
- Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Chengdu, China
| | - Yongtao Li
- Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Jun Li
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, China
| | - Yu Chen
- Institute of Nanjiang Yellow Goat Sciences, Bazhong, Sichuan, China
| | - Hengbo Shi
- Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, Hangzhou, China
| |
Collapse
|
50
|
Real MVF, Colvin MS, Sheehan MJ, Moeller AH. Major urinary protein ( Mup) gene family deletion drives sex-specific alterations in the house-mouse gut microbiota. Microbiol Spectr 2024; 12:e0356623. [PMID: 38170981 PMCID: PMC10846032 DOI: 10.1128/spectrum.03566-23] [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: 10/04/2023] [Accepted: 11/23/2023] [Indexed: 01/05/2024] Open
Abstract
The gut microbiota is shaped by host metabolism. In house mice (Mus musculus), major urinary protein (MUP) pheromone production represents a considerable energy investment, particularly in sexually mature males. Deletion of the Mup gene family shifts mouse metabolism toward an anabolic state, marked by lipogenesis, lipid accumulation, and body mass increases. Given the metabolic implications of MUPs, they may also influence the gut microbiota. Here, we investigated the effect of a deletion of the Mup gene family on the gut microbiota of sexually mature mice. Shotgun metagenomics revealed distinct taxonomic and functional profiles between wild-type and knockout males but not females. Deletion of the Mup gene cluster significantly reduced diversity in microbial families and functions in male mice. Additionally, a species of Ruminococcaceae and several microbial functions, such as transporters involved in vitamin B5 acquisition, were significantly depleted in the microbiota of Mup knockout males. Altogether, these results show that MUPs significantly affect the gut microbiota of house mouse in a sex-specific manner.IMPORTANCEThe community of microorganisms that inhabits the gastrointestinal tract can have profound effects on host phenotypes. The gut microbiota is in turn shaped by host genes, including those involved with host metabolism. In adult male house mice, expression of the major urinary protein (Mup) gene cluster represents a substantial energy investment, and deletion of the Mup gene family leads to fat accumulation and weight gain in males. We show that deleting Mup genes also alters the gut microbiota of male, but not female, mice in terms of both taxonomic and functional compositions. Male mice without Mup genes harbored fewer gut bacterial families and reduced abundance of a species of Ruminococcaceae, a family that has been previously shown to reduce obesity risk. Studying the impact of the Mup gene family on the gut microbiota has the potential to reveal the ways in which these genes affect host phenotypes.
Collapse
Affiliation(s)
- Madalena V. F. Real
- Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, New York, USA
| | - Melanie S. Colvin
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA
| | - Michael J. Sheehan
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA
| | - Andrew H. Moeller
- Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, New York, USA
| |
Collapse
|