1
|
Usuda D, Sugita M, Shen P, Umehara T, Kitamoto T. Leaky Gut Syndrome Along With Clostridium perfringens Bacteremia in a Neurodegenerative Disease Patient: A Case Report. Cureus 2024; 16:e75290. [PMID: 39776697 PMCID: PMC11703780 DOI: 10.7759/cureus.75290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2024] [Indexed: 01/11/2025] Open
Abstract
Leaky gut syndrome (LGS) is caused by intestinal epithelial injury and increased intestinal permeability due to a variety of factors, including chronic stress, inflammatory bowel disease, diabetes, surgery, and chemotherapy, resulting in an increased influx of matter from the intestinal lumen causing constipation and bacteremia. To our knowledge, this is the first known case of LGS along with Clostridium perfringens (C. perfringens) bacteremia in a neurodegenerative disease patient. The patient was an 81-year-old male with a history of Alzheimer's disease, cerebral infarction, and diverticulitis in a psychiatric hospital, fed via a nasogastric tube. During hospitalization, he developed a 37.4℃ temperature and disturbance of consciousness evaluated as 3 points on the Glasgow Coma Scale. A follow-up blood examination revealed a white blood cell count of 29,000/µL and a C-reactive protein value of 11.2 mg/dL. Computed tomography revealed an increased concentration of peripheral adipose tissue from the sigmoid colon to the rectum and significant quantities of stool in the rectum. Treatment was initiated with doripenem (DRPM) for sepsis of unknown focus. C. perfringens was subsequently identified in both two blood culture tests. He improved with decreased inflammatory response; DRPM was terminated after 14 days. He remains free of recurrence. We speculate that the LGS in this case may have developed from dopaminergic neuronal decrease and impaired amino acid metabolism caused by chronic hypo-inflammation due to neurodegenerative disease (Alzheimer's disease). We report the first known case of LGS along with C. perfringens bacteremia in a neurodegenerative disease patient.
Collapse
Affiliation(s)
- Daisuke Usuda
- Department of Emergency and Critical Care Medicine, Juntendo University Nerima Hospital, Nerima, JPN
- Department of Internal Medicine, Hasegawa Hospital, Mitaka, JPN
| | - Manabu Sugita
- Department of Emergency and Critical Care Medicine, Juntendo University Nerima Hospital, Nerima, JPN
- Department of Internal Medicine, Hasegawa Hospital, Mitaka, JPN
| | - Pingcheng Shen
- Department of Internal Medicine, Hasegawa Hospital, Mitaka, JPN
| | - Tadashi Umehara
- Department of Internal Medicine, Hasegawa Hospital, Mitaka, JPN
| | | |
Collapse
|
2
|
Damanti S, Senini E, De Lorenzo R, Merolla A, Santoro S, Festorazzi C, Messina M, Vitali G, Sciorati C, Rovere-Querini P. Acute Sarcopenia: Mechanisms and Management. Nutrients 2024; 16:3428. [PMID: 39458423 PMCID: PMC11510680 DOI: 10.3390/nu16203428] [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: 08/26/2024] [Revised: 09/27/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Acute sarcopenia refers to the swift decline in muscle function and mass following acute events such as illness, surgery, trauma, or burns that presents significant challenges in hospitalized older adults. METHODS narrative review to describe the mechanisms and management of acute sarcopenia. RESULTS The prevalence of acute sarcopenia ranges from 28% to 69%, likely underdiagnosed due to the absence of muscle mass and function assessments in most clinical settings. Systemic inflammation, immune-endocrine dysregulation, and anabolic resistance are identified as key pathophysiological factors. Interventions include early mobilization, resistance exercise, neuromuscular electrical stimulation, and nutritional strategies such as protein supplementation, leucine, β-hydroxy-β-methyl-butyrate, omega-3 fatty acids, and creatine monohydrate. Pharmaceuticals show variable efficacy. CONCLUSIONS Future research should prioritize serial monitoring of muscle parameters, identification of predictive biomarkers, and the involvement of multidisciplinary teams from hospital admission to address sarcopenia. Early and targeted interventions are crucial to improve outcomes and prevent long-term disability associated with acute sarcopenia.
Collapse
Affiliation(s)
- Sarah Damanti
- Internal Medicine Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (S.D.); (G.V.); (P.R.-Q.)
- Division of Immunology, Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, 20100 Milan, Italy; (E.S.); (R.D.L.); (A.M.); (S.S.); (C.F.); (M.M.)
| | - Eleonora Senini
- Division of Immunology, Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, 20100 Milan, Italy; (E.S.); (R.D.L.); (A.M.); (S.S.); (C.F.); (M.M.)
| | - Rebecca De Lorenzo
- Division of Immunology, Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, 20100 Milan, Italy; (E.S.); (R.D.L.); (A.M.); (S.S.); (C.F.); (M.M.)
| | - Aurora Merolla
- Division of Immunology, Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, 20100 Milan, Italy; (E.S.); (R.D.L.); (A.M.); (S.S.); (C.F.); (M.M.)
| | - Simona Santoro
- Division of Immunology, Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, 20100 Milan, Italy; (E.S.); (R.D.L.); (A.M.); (S.S.); (C.F.); (M.M.)
| | - Costanza Festorazzi
- Division of Immunology, Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, 20100 Milan, Italy; (E.S.); (R.D.L.); (A.M.); (S.S.); (C.F.); (M.M.)
| | - Marco Messina
- Division of Immunology, Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, 20100 Milan, Italy; (E.S.); (R.D.L.); (A.M.); (S.S.); (C.F.); (M.M.)
| | - Giordano Vitali
- Internal Medicine Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (S.D.); (G.V.); (P.R.-Q.)
| | - Clara Sciorati
- Division of Immunology, Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, 20100 Milan, Italy; (E.S.); (R.D.L.); (A.M.); (S.S.); (C.F.); (M.M.)
| | - Patrizia Rovere-Querini
- Internal Medicine Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (S.D.); (G.V.); (P.R.-Q.)
- Division of Immunology, Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, 20100 Milan, Italy; (E.S.); (R.D.L.); (A.M.); (S.S.); (C.F.); (M.M.)
| |
Collapse
|
3
|
Csiszar A, Ungvari A, Patai R, Gulej R, Yabluchanskiy A, Benyo Z, Kovacs I, Sotonyi P, Kirkpartrick AC, Prodan CI, Liotta EM, Zhang XA, Toth P, Tarantini S, Sorond FA, Ungvari Z. Atherosclerotic burden and cerebral small vessel disease: exploring the link through microvascular aging and cerebral microhemorrhages. GeroScience 2024; 46:5103-5132. [PMID: 38639833 PMCID: PMC11336042 DOI: 10.1007/s11357-024-01139-7] [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: 02/20/2024] [Accepted: 03/14/2024] [Indexed: 04/20/2024] Open
Abstract
Cerebral microhemorrhages (CMHs, also known as cerebral microbleeds) are a critical but frequently underestimated aspect of cerebral small vessel disease (CSVD), bearing substantial clinical consequences. Detectable through sensitive neuroimaging techniques, CMHs reveal an extensive pathological landscape. They are prevalent in the aging population, with multiple CMHs often being observed in a given individual. CMHs are closely associated with accelerated cognitive decline and are increasingly recognized as key contributors to the pathogenesis of vascular cognitive impairment and dementia (VCID) and Alzheimer's disease (AD). This review paper delves into the hypothesis that atherosclerosis, a prevalent age-related large vessel disease, extends its pathological influence into the cerebral microcirculation, thereby contributing to the development and progression of CSVD, with a specific focus on CMHs. We explore the concept of vascular aging as a continuum, bridging macrovascular pathologies like atherosclerosis with microvascular abnormalities characteristic of CSVD. We posit that the same risk factors precipitating accelerated aging in large vessels (i.e., atherogenesis), primarily through oxidative stress and inflammatory pathways, similarly instigate accelerated microvascular aging. Accelerated microvascular aging leads to increased microvascular fragility, which in turn predisposes to the formation of CMHs. The presence of hypertension and amyloid pathology further intensifies this process. We comprehensively overview the current body of evidence supporting this interconnected vascular hypothesis. Our review includes an examination of epidemiological data, which provides insights into the prevalence and impact of CMHs in the context of atherosclerosis and CSVD. Furthermore, we explore the shared mechanisms between large vessel aging, atherogenesis, microvascular aging, and CSVD, particularly focusing on how these intertwined processes contribute to the genesis of CMHs. By highlighting the role of vascular aging in the pathophysiology of CMHs, this review seeks to enhance the understanding of CSVD and its links to systemic vascular disorders. Our aim is to provide insights that could inform future therapeutic approaches and research directions in the realm of neurovascular health.
Collapse
Affiliation(s)
- Anna Csiszar
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Ungvari
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary.
| | - Roland Patai
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Rafal Gulej
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Zoltan Benyo
- Institute of Translational Medicine, Semmelweis University, 1094, Budapest, Hungary
- Cerebrovascular and Neurocognitive Disorders Research Group, HUN-REN, Semmelweis University, 1094, Budapest, Hungary
| | - Illes Kovacs
- Department of Ophthalmology, Semmelweis University, 1085, Budapest, Hungary
- Department of Ophthalmology, Weill Cornell Medical College, New York, NY, 10021, USA
| | - Peter Sotonyi
- Department of Vascular and Endovascular Surgery, Heart and Vascular Centre, Semmelweis University, 1122, Budapest, Hungary
| | - Angelia C Kirkpartrick
- Veterans Affairs Medical Center, Oklahoma City, OK, USA
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Calin I Prodan
- Veterans Affairs Medical Center, Oklahoma City, OK, USA
- Department of Neurology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Eric M Liotta
- International Training Program in Geroscience, Doctoral College/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Neurology, Division of Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Xin A Zhang
- Department of Physiology, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Peter Toth
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary
- Department of Neurosurgery, Medical School, University of Pecs, Pecs, Hungary
- Neurotrauma Research Group, Szentagothai Research Centre, University of Pecs, Pecs, Hungary
- ELKH-PTE Clinical Neuroscience MR Research Group, University of Pecs, Pecs, Hungary
| | - Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Farzaneh A Sorond
- Department of Neurology, Division of Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Department of Public Health, Semmelweis University, Budapest, Hungary
| |
Collapse
|
4
|
Monnig M, Shah K. Linking alcohol use to Alzheimer's disease: Interactions with aging and APOE along immune pathways. MEDICAL RESEARCH ARCHIVES 2024; 12:10.18103/mra.v12i8.5228. [PMID: 39544182 PMCID: PMC11563488 DOI: 10.18103/mra.v12i8.5228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Although it is known that APOE genotype is the strongest genetic risk factor for late-onset Alzheimer's disease, development is a multifactorial process. Alcohol use is a contributor to the epidemic of Alzheimer's disease and related dementias in the US and globally, yet mechanisms are not fully understood. Carriers of the APOE ε4 allele show elevated risk of dementia in relation to several lifestyle factors, including alcohol use. In this review, we describe how alcohol interacts with APOE genotype and aging with potential implications for Alzheimer's disease promotion. Age-related immune senescence and "inflammaging" (i.e., low-grade inflammation associated with aging) are increasingly recognized as contributors to age-related disease. We focus on three immune pathways that are likely contributors to Alzheimer's disease development, centering on alcohol and APOE genotype interactions, specifically: 1) microbial translocation and immune activation, 2) the senescence associated secretory phenotype, and 3) neuroinflammation. First, microbial translocation, the unphysiological movement of gut products into systemic circulation, elicits a proinflammatory response and increases with aging, with proposed links to Alzheimer's disease. Second, the senescence associated secretory phenotype is a set of intercellular signaling factors, e.g., proinflammatory cytokines and chemokines, growth regulators, and proteases, that drives cellular aging when senescent cells remain metabolically active. The senescence associated secretory phenotype can drive development of aging-diseases such as Alzheimer's disease. Third, neuroinflammation occurs via numerous mechanisms such as microglial activation and is gaining recognition as an etiological factor in the development of Alzheimer's disease. This review focuses on interactions of alcohol with APOE genotype and aging along these three pathways that may promote Alzheimer's disease. Further research on these processes may inform development of strategies to prevent onset and progression of Alzheimer's disease and to delay associated cognitive decline.
Collapse
Affiliation(s)
- Mollie Monnig
- Center for Alcohol and Addiction Studies, Brown University, Providence, RI 02912, USA
| | - Krish Shah
- Center for Alcohol and Addiction Studies, Brown University, Providence, RI 02912, USA
| |
Collapse
|
5
|
Maher SA, AbdAllah NB, Ageeli EA, Riad E, Kattan SW, Abdelaal S, Abdelfatah W, Ibrahim GA, Toraih EA, Awadalla GA, Fawzy MS, Ibrahim A. Impact of Interleukin-17 Receptor A Gene Variants on Asthma Susceptibility and Clinical Manifestations in Children and Adolescents. CHILDREN (BASEL, SWITZERLAND) 2024; 11:657. [PMID: 38929236 PMCID: PMC11202101 DOI: 10.3390/children11060657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/24/2024] [Accepted: 05/26/2024] [Indexed: 06/28/2024]
Abstract
Several single nucleotide polymorphisms (SNPs) in multiple interleukin receptor genes could be associated with asthma risk and/or phenotype. Interleukin-17 (IL-17) has been implicated in tissue inflammation and autoimmune diseases. As no previous studies have uncovered the potential role of IL17 receptor A (RA) gene variants in asthma risk, we aimed to explore the association of four IL17RA SNPs (i.e., rs4819554A/G, rs879577C/T, rs41323645G/A, and rs4819555C/T) with asthma susceptibility/phenotype in our region. TaqMan allelic discrimination analysis was used to genotype 192 individuals. We found that the rs4819554 G/G genotype significantly reduced disease risk in the codominant (OR = 0.15, 95%CI = 0.05-0.45, p < 0.001), dominant (OR = 0.49, 95%CI = 0.26-0.93, p = 0.028), and recessive (OR = 0.18, 95%CI = 0.07-0.52, p < 0.001) models. Similarly, rs879577 showed reduced disease risk associated with the T allele across all genetic models. However, the A allele of rs41323645 was associated with increased disease risk in all models. The G/A and A/A genotypes have higher ORs of 2.47 (95%CI = 1.19-5.14) and 3.86 (95%CI = 1.62-9.18), respectively. Similar trends are observed in the dominant 2.89 (95%CI = 1.47-5.68, p = 0.002) and recessive 2.34 (95%CI = 1.10-4.98, p = 0.025) models. For the rs4819555 variant, although there was no significant association identified under any models, carriers of the rs4819554*A demonstrated an association with a positive family history of asthma (71.4% in carriers vs. 27% in non-carriers; p = 0.025) and the use of relievers for >2 weeks (52.2% of carriers vs. 28.8% of non-carriers; p = 0.047). Meanwhile, the rs4819555*C carriers displayed a significant divergence in the asthma phenotype, specifically atopic asthma (83.3% vs. 61.1%; p = 0.007), showed a higher prevalence of chest tightness (88.9% vs. 61.5%; p = 0.029), and were more likely to report comorbidities (57.7% vs. 16.7%, p = 0.003). The most frequent haplotype in the asthma group was ACAC, with a frequency of 22.87% vs. 1.36% in the controls (p < 0.001). In conclusion, the studied IL17RA variants could be essential in asthma susceptibility and phenotype in children and adolescents.
Collapse
Affiliation(s)
- Shymaa Ahmed Maher
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt;
- Center of Excellence in Molecular and Cellular Medicine (CEMCM), Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Nouran B. AbdAllah
- Department of Pediatrics, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt; (N.B.A.); (S.A.); (A.I.)
| | - Essam Al Ageeli
- Department of Basic Medical Sciences, Faculty of Medicine, Jazan University, Jazan 45141, Saudi Arabia;
| | - Eman Riad
- Department of Chest Diseases and Tuberculosis, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt; (E.R.); (W.A.)
| | - Shahad W. Kattan
- Department of Medical Laboratory, College of Applied Medical Sciences, Taibah University, Yanbu 46423, Saudi Arabia;
| | - Sherouk Abdelaal
- Department of Pediatrics, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt; (N.B.A.); (S.A.); (A.I.)
| | - Wagdy Abdelfatah
- Department of Chest Diseases and Tuberculosis, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt; (E.R.); (W.A.)
| | - Gehan A. Ibrahim
- Department of Clinical Pathology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt;
| | - Eman A. Toraih
- Department of Surgery, School of Medicine, Tulane University, New Orleans, LA 70112, USA;
- Medical Genetics Unit, Department of Histology and Cell Biology, Suez Canal University, Ismailia 41522, Egypt
| | - Ghada A. Awadalla
- Biochemistry Department, Animal Health Research Institute, Mansoura Branch, Giza 12618, Egypt;
| | - Manal S. Fawzy
- Department of Biochemistry, Faculty of Medicine, Northern Border University, Arar P.O. Box 1321, Saudi Arabia
| | - Ahmed Ibrahim
- Department of Pediatrics, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt; (N.B.A.); (S.A.); (A.I.)
| |
Collapse
|
6
|
Boyajian JL, Islam P, Abosalha A, Schaly S, Thareja R, Kassab A, Arora K, Santos M, Shum-Tim C, Prakash S. Probiotics, prebiotics, synbiotics and other microbiome-based innovative therapeutics to mitigate obesity and enhance longevity via the gut-brain axis. MICROBIOME RESEARCH REPORTS 2024; 3:29. [PMID: 39421246 PMCID: PMC11480732 DOI: 10.20517/mrr.2024.05] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/18/2024] [Accepted: 05/11/2024] [Indexed: 10/19/2024]
Abstract
The global prevalence of obesity currently exceeds 1 billion people and is accompanied by an increase in the aging population. Obesity and aging share many hallmarks and are leading risk factors for cardiometabolic disease and premature death. Current anti-obesity and pro-longevity pharmacotherapies are limited by side effects, warranting the development of novel therapies. The gut microbiota plays a major role in human health and disease, with a dysbiotic composition evident in obese and aged individuals. The bidirectional communication system between the gut and the central nervous system, known as the gut-brain axis, may link obesity to unhealthy aging. Modulating the gut with microbiome-targeted therapies, such as biotics, is a novel strategy to treat and/or manage obesity and promote longevity. Biotics represent material derived from living or once-living organisms, many of which have therapeutic effects. Pre-, pro-, syn- and post-biotics may beneficially modulate gut microbial composition and function to improve obesity and the aging process. However, the investigation of biotics as next-generation therapeutics has only just begun. Further research is needed to identify therapeutic biotics and understand their mechanisms of action. Investigating the function of the gut-brain axis in obesity and aging may lead to novel therapeutic strategies for obese, aged and comorbid (e.g., sarcopenic obese) patient populations. This review discusses the interrelationship between obesity and aging, with a particular emphasis on the gut microbiome, and presents biotics as novel therapeutic agents for obesity, aging and related disease states.
Collapse
Affiliation(s)
- Jacqueline L. Boyajian
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Paromita Islam
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Ahmed Abosalha
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
- Pharmaceutical Technology Department, Faculty of Pharmacy, Tanta University, Tanta 31111, Egypt
| | - Sabrina Schaly
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Rahul Thareja
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Amal Kassab
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Karan Arora
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Madison Santos
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Cedrique Shum-Tim
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Satya Prakash
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| |
Collapse
|
7
|
Yang W, Xi C, Yao H, Yuan Q, Zhang J, Chen Q, Wu G, Hu J. Oral administration of lysozyme protects against injury of ileum via modulating gut microbiota dysbiosis after severe traumatic brain injury. Front Cell Infect Microbiol 2024; 14:1304218. [PMID: 38352055 PMCID: PMC10861676 DOI: 10.3389/fcimb.2024.1304218] [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: 09/29/2023] [Accepted: 01/16/2024] [Indexed: 02/16/2024] Open
Abstract
Objective The current study sought to clarify the role of lysozyme-regulated gut microbiota and explored the potential therapeutic effects of lysozyme on ileum injury induced by severe traumatic brain injury (sTBI) and bacterial pneumonia in vivo and in vitro experiments. Methods Male 6-8-week-old specific pathogen-free (SPF) C57BL/6 mice were randomly divided into Normal group (N), Sham group (S), sTBI group (T), sTBI + or Lysozyme-treated group (L), Normal + Lysozyme group (NL) and Sham group + Lysozyme group (SL). At the day 7 after establishment of the model, mice were anesthetized and the samples were collected. The microbiota in lungs and fresh contents of the ileocecum were analyzed. Lungs and distal ileum were used to detect the degree of injury. The number of Paneth cells and the expression level of lysozyme were assessed. The bacterial translocation was determined. Intestinal organoids culture and co-coculture system was used to test whether lysozyme remodels the intestinal barrier through the gut microbiota. Results After oral administration of lysozyme, the intestinal microbiota is rebalanced, the composition of lung microbiota is restored, and translocation of intestinal bacteria is mitigated. Lysozyme administration reinstates lysozyme expression in Paneth cells, thereby reducing intestinal permeability, pathological score, apoptosis rate, and inflammation levels. The gut microbiota, including Oscillospira, Ruminococcus, Alistipes, Butyricicoccus, and Lactobacillus, play a crucial role in regulating and improving intestinal barrier damage and modulating Paneth cells in lysozyme-treated mice. A co-culture system comprising intestinal organoids and brain-derived proteins (BP), which demonstrated that the BP effectively downregulated the expression of lysozyme in intestinal organoids. However, supplementation of lysozyme to this co-culture system failed to restore its expression in intestinal organoids. Conclusion The present study unveiled a virtuous cycle whereby oral administration of lysozyme restores Paneth cell's function, mitigates intestinal injury and bacterial translocation through the remodeling of gut microbiota.
Collapse
Affiliation(s)
- Weijian Yang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Caihua Xi
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Department of Neurosurgery and Neurocritical Care, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Haijun Yao
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Department of Neurosurgery and Neurocritical Care, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qiang Yuan
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Jun Zhang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Qifang Chen
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Department of Neurosurgery and Neurocritical Care, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Gang Wu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Jin Hu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| |
Collapse
|
8
|
Ureña E, Xu B, Regan JC, Atilano ML, Minkley LJ, Filer D, Lu YX, Bolukbasi E, Khericha M, Alic N, Partridge L. Trametinib ameliorates aging-associated gut pathology in Drosophila females by reducing Pol III activity in intestinal stem cells. Proc Natl Acad Sci U S A 2024; 121:e2311313121. [PMID: 38241436 PMCID: PMC10823232 DOI: 10.1073/pnas.2311313121] [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: 07/12/2023] [Accepted: 11/17/2023] [Indexed: 01/21/2024] Open
Abstract
Pharmacological therapies are promising interventions to slow down aging and reduce multimorbidity in the elderly. Studies in animal models are the first step toward translation of candidate molecules into human therapies, as they aim to elucidate the molecular pathways, cellular mechanisms, and tissue pathologies involved in the anti-aging effects. Trametinib, an allosteric inhibitor of MEK within the Ras/MAPK (Ras/Mitogen-Activated Protein Kinase) pathway and currently used as an anti-cancer treatment, emerged as a geroprotector candidate because it extended lifespan in the fruit fly Drosophila melanogaster. Here, we confirm that trametinib consistently and robustly extends female lifespan, and reduces intestinal stem cell (ISC) proliferation, tumor formation, tissue dysplasia, and barrier disruption in guts in aged flies. In contrast, pro-longevity effects of trametinib are weak and inconsistent in males, and it does not influence gut homeostasis. Inhibition of the Ras/MAPK pathway specifically in ISCs is sufficient to partially recapitulate the effects of trametinib. Moreover, in ISCs, trametinib decreases the activity of the RNA polymerase III (Pol III), a conserved enzyme synthesizing transfer RNAs and other short, non-coding RNAs, and whose inhibition also extends lifespan and reduces gut pathology. Finally, we show that the pro-longevity effect of trametinib in ISCs is partially mediated by Maf1, a repressor of Pol III, suggesting a life-limiting Ras/MAPK-Maf1-Pol III axis in these cells. The mechanism of action described in this work paves the way for further studies on the anti-aging effects of trametinib in mammals and shows its potential for clinical application in humans.
Collapse
Affiliation(s)
- Enric Ureña
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Bowen Xu
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Jennifer C. Regan
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Magda L. Atilano
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Lucy J. Minkley
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Danny Filer
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Yu-Xuan Lu
- Max Planck Institute for Biology of Ageing, CologneD-50931, Germany
| | - Ekin Bolukbasi
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Mobina Khericha
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Nazif Alic
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Linda Partridge
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
- Max Planck Institute for Biology of Ageing, CologneD-50931, Germany
| |
Collapse
|
9
|
Réus GZ, Manosso LM, Quevedo J, Carvalho AF. Major depressive disorder as a neuro-immune disorder: Origin, mechanisms, and therapeutic opportunities. Neurosci Biobehav Rev 2023; 155:105425. [PMID: 37852343 DOI: 10.1016/j.neubiorev.2023.105425] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/16/2023] [Accepted: 10/12/2023] [Indexed: 10/20/2023]
Abstract
Notwithstanding advances in understanding the pathophysiology of major depressive disorder (MDD), no single mechanism can explain all facets of this disorder. An expanding body of evidence indicates a putative role for the inflammatory response. Several meta-analyses showed an increase in systemic peripheral inflammatory markers in individuals with MDD. Numerous conditions and circumstances in the modern world may promote chronic systemic inflammation through mechanisms, including alterations in the gut microbiota. Peripheral cytokines may reach the brain and contribute to neuroinflammation through cellular, humoral, and neural pathways. On the other hand, antidepressant drugs may decrease peripheral levels of inflammatory markers. Anti-inflammatory drugs and nutritional strategies that reduce inflammation also could improve depressive symptoms. The present study provides a critical review of recent advances in the role of inflammation in the pathophysiology of MDD. Furthermore, this review discusses the role of glial cells and the main drivers of changes associated with neuroinflammation. Finally, we highlight possible novel neurotherapeutic targets for MDD that could exert antidepressant effects by modulating inflammation.
Collapse
Affiliation(s)
- Gislaine Z Réus
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| | - Luana M Manosso
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - João Quevedo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil; Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - André F Carvalho
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| |
Collapse
|
10
|
Kuai Z, Chao X, He Y, Ren W. Metformin attenuates inflammation and boosts autophagy in the liver and intestine of chronologically aged rats. Exp Gerontol 2023; 184:112331. [PMID: 37967593 DOI: 10.1016/j.exger.2023.112331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 11/09/2023] [Accepted: 11/12/2023] [Indexed: 11/17/2023]
Abstract
BACKGROUND Our previous studies found that autophagy levels in liver and intestinal segments of naturally aging rats were downregulated, and the expression of pro-inflammatory factors was increased. The increased expression of pro-inflammatory factors might be related to the downregulation of autophagy. AMPK is the most critical upstream targeting and regulating molecule of autophagy, and Metformin, as an agonist of AMPK, has the effects of anti-inflammation and anti-aging. We pretreated 29-month-old naturally aging rats with Metformin for a short period and observed the changes in autophagy levels and pro-inflammatory factors in the liver, ileum, and colon after 31 days of intervention and preliminarily investigated the mechanism of its action. METHODS 29-month-old SPF male Wistar rats were divided into three groups: The control group, the Metformin 100 mg/kg intervention group, and the Metformin 250 mg/kg intervention group, with eight rats in each group. At 29 months, different concentrations of Metformin (100 mg/kg, 250 mg/kg) were given by gavage once a day until 30 months, and the control group was kept generally until 30 months. Western Blot was used to assess the expression levels of AMPK, P-AMPK, LC3, and P62 proteins in the liver and intestinal tissues. Intestinal and liver tissues were immunofluorescence labeled for LC3 and P62 proteins. Moreover, RT-qPCR was conducted to detect the expression levels of pro-inflammatory factors IL-1β, TNF-α, IL-6, and MMP-9 mRNA in liver and intestinal tissues. RESULTS Short-term Metformin pretreatment (31 days) in naturally aging rats (29 months old) increased autophagy levels and down-regulated the expression of various pro-inflammatory cytokines (IL-1β, TNF-α, MMP-9, and IL-6) in various intestinal segments and the liver-the expression of LC3II protein enriched with the increase of Metformin concentration. The level of P62 protein decreased with the accumulation of Metformin concentration. And a higher concentration of Metformin was associated with increased expression of P-AMPK protein. CONCLUSIONS Metformin intervention can boost the autophagy level in the liver and intestine and reduce the expression of aging-related inflammatory factors in aged rats, and these effects may be related to the increase of the AMPK phosphorylation level.
Collapse
Affiliation(s)
- Zheng Kuai
- Department of Geriatrics, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xin Chao
- Department of Geriatrics, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yuting He
- Department of Geriatrics, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Weiying Ren
- Department of Geriatrics, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
11
|
Kim CS, Jung MH, Choi EY, Shin DM. Probiotic supplementation has sex-dependent effects on immune responses in association with the gut microbiota in community-dwelling older adults: a randomized, double-blind, placebo-controlled, multicenter trial. Nutr Res Pract 2023; 17:883-898. [PMID: 37780220 PMCID: PMC10522805 DOI: 10.4162/nrp.2023.17.5.883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 05/13/2023] [Accepted: 05/19/2023] [Indexed: 10/03/2023] Open
Abstract
BACKGROUND/OBJECTIVES Probiotics have been suggested as potent modulators of age-related disorders in immunological functions, yet little is known about sex-dependent effects of probiotic supplements. Therefore, we aimed to investigate sex-dependent effects of probiotics on profiles of the gut microbiota and peripheral immune cells in healthy older adults. SUBJECTS/METHODS In a randomized, double-blind, placebo-controlled, multicenter trial, healthy elderly individuals ≥ 65 yrs old were administered probiotic capsules (or placebo) for 12 wk. Gut microbiota was analyzed using 16S rRNA gene sequencing and bioinformatic analyses. Peripheral immune cells were profiled using flow cytometry for lymphocytes (natural killer, B, CD4+ T, and CD8+ T cells), dendritic cells, monocytes, and their subpopulations. RESULTS Compared with placebo, phylum Firmicutes was significantly reduced in the probiotic group in women, but not in men. At the genus level, sex-specific responses included reductions in the relative abundances of pro-inflammatory gut microbes, including Catabacter and unclassified_Coriobacteriales, and Burkholderia and unclassified Enterobacteriaceae, in men and women, respectively. Peripheral immune cell profiling analysis revealed that in men, probiotics significantly reduced the proportions of dendritic cells and CD14+ CD16- monocytes; however, these effects were not observed in women. In contrast, the proportion of total CD4+ T cells was significantly reduced in women in the probiotic group. Additionally, serum lipopolysaccharide-binding protein levels showed a decreasing tendency that were positively associated with changes in gut bacteria, including Catabacter (ρ = 0.678, P < 0.05) and Burkholderia (ρ = 0.673, P < 0.05) in men and women, respectively. CONCLUSIONS These results suggest that probiotic supplementation may reduce the incidence of inflammation-related diseases by regulating the profiles of the gut microbiota and peripheral immune cells in healthy elders in a sex-specific manner.
Collapse
Affiliation(s)
- Chong-Su Kim
- Department of Food and Nutrition, College of Natural Information Sciences, Dongduk Women's University, Seoul 02748, Korea
| | - Min Ho Jung
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Eun Young Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Dong-Mi Shin
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
12
|
Vollrath S, Matits L, Jerg A, Zorn J, John L, Steinacker JM, Bizjak DA. Blood Profiling of Athletes after COVID-19: Differences in Blood Profiles of Post-COVID-19 Athletes Compared to Uninfected Athletic Individuals-An Exploratory Analysis. Biomedicines 2023; 11:1911. [PMID: 37509550 PMCID: PMC10377547 DOI: 10.3390/biomedicines11071911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/29/2023] [Accepted: 07/02/2023] [Indexed: 07/30/2023] Open
Abstract
Blood profiling data in athletic populations and their respective responses to SARS-CoV-2 infection are lacking. Thus, this exploratory pilot study aimed to analyze and compare clinical blood markers in previously infected trained athletes (ATH; 30 m/29 f) and a not previously infected healthy athletic control group (HC; 12 m/19 f). The ATH group undertook a sports medical examination which included extended blood analyses. Blood profiles with a total of 74 variables were assessed (blood counts, pro-/inflammatory and immunological markers, and micronutrients), and the ATH group was compared to the age-matched, vaccinated HC group with comparable athletic back grounds, though without previous SARS-CoV-2-infections. The ATH group showed lower IgG, Troponin-T levels, and they had a lower complement/acute-phase protein activation. Furthermore, Vitamin D levels were lower and electrolyte/micronutrient concentrations were higher in ATH. Soluble transferrin receptor as a marker of erythrocyte turnover was decreased whereas PTT as a coagulation marker was increased. Subgroup analyses according to sex revealed more differences between the women of the ATH and HC groups (for 25 different variables) than between the men (for 5 different variables), especially for immunological and metabolic variables. In particular, the immune system and electrolyte/micronutrient status should be observed frequently and sex-specifically in this athletic cohort.
Collapse
Affiliation(s)
- Shirin Vollrath
- Division of Sports and Rehabilitation Medicine, Department of Medicine, Ulm University Hospital, 89075 Ulm, Germany
| | - Lynn Matits
- Division of Sports and Rehabilitation Medicine, Department of Medicine, Ulm University Hospital, 89075 Ulm, Germany
- Division of Clinical & Biological Psychology, Institute of Psychology and Education, Ulm University, 89075 Ulm, Germany
| | - Achim Jerg
- Division of Sports and Rehabilitation Medicine, Department of Medicine, Ulm University Hospital, 89075 Ulm, Germany
| | - Jule Zorn
- Division of Sports and Rehabilitation Medicine, Department of Medicine, Ulm University Hospital, 89075 Ulm, Germany
| | - Lucas John
- Division of Sports and Rehabilitation Medicine, Department of Medicine, Ulm University Hospital, 89075 Ulm, Germany
| | - Jürgen Michael Steinacker
- Division of Sports and Rehabilitation Medicine, Department of Medicine, Ulm University Hospital, 89075 Ulm, Germany
| | - Daniel Alexander Bizjak
- Division of Sports and Rehabilitation Medicine, Department of Medicine, Ulm University Hospital, 89075 Ulm, Germany
| |
Collapse
|
13
|
Chittimalli K, Jahan J, Sakamuri A, McAdams ZL, Ericsson AC, Jarajapu YP. Restoration of the gut barrier integrity and restructuring of the gut microbiome in aging by angiotensin-(1-7). Clin Sci (Lond) 2023; 137:913-930. [PMID: 37254732 PMCID: PMC10881191 DOI: 10.1042/cs20220904] [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: 01/03/2023] [Revised: 05/16/2023] [Accepted: 05/30/2023] [Indexed: 06/01/2023]
Abstract
Compromised barrier function of colon epithelium with aging is largely due to gut microbial dysbiosis. Recent studies implicate an important role for angiotensin converting enzymes, ACE and ACE2, angiotensins, and the receptors, AT1 receptor (AT1R) and Mas receptor (MasR), in the regulation of colon functions. The present study tested the hypothesis that leaky gut in aging is associated with an imbalance in ACE2/ACE and that the treatment with angiotenisn-(1-7) (Ang-(1-7)) will restore gut barrier integrity and microbiome. Studies were carried out in Young (3-4 months) and old (20-24 months) male mice. Ang-(1-7) was administered by using osmotic pumps. Outcome measures included expressions of ACE, ACE2, AT1R, and MasR, intestinal permeability by using FITC-dextran, and immunohistochemistry of claudin 1 and occludin, and intestinal stem cells (ISCs). ACE2 protein and activity were decreased in Old group while that of ACE were unchanged. Increased intestinal permeability and plasma levels of zonulin-1 in the Old group were normalized by Ang-(1-7). Epithelial disintegrity, reduced number of goblet cells and ISCs in the old group were restored by Ang-(1-7). Expression of claudin 1 and occludin in the aging colon was increased by Ang-(1-7). Infiltration of CD11b+ or F4/80+ inflammatory cells in the old colons were decreased by Ang-(1-7). Gut microbial dysbiosis in aging was evident by decreased richness and altered beta diversity that were reversed by Ang-(1-7) with increased abundance of Lactobacillus or Lachnospiraceae. The present study shows that Ang-(1-7) restores gut barrier integrity and reduces inflammation in the aging colon by restoring the layer of ISCs and by restructuring the gut microbiome.
Collapse
Affiliation(s)
- Kishore Chittimalli
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Fargo, ND, U.S.A
| | - Jesmin Jahan
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Fargo, ND, U.S.A
| | - Anil Sakamuri
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Fargo, ND, U.S.A
| | - Zachary L. McAdams
- Missouri Metagenomics Center, Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, U.S.A
| | - Aaron C. Ericsson
- Missouri Metagenomics Center, Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, U.S.A
| | - Yagna P.R. Jarajapu
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Fargo, ND, U.S.A
| |
Collapse
|
14
|
Salazar AM, Aparicio R, Clark RI, Rera M, Walker DW. Intestinal barrier dysfunction: an evolutionarily conserved hallmark of aging. Dis Model Mech 2023; 16:dmm049969. [PMID: 37144684 PMCID: PMC10184675 DOI: 10.1242/dmm.049969] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023] Open
Abstract
A major challenge in the biology of aging is to understand how specific age-onset pathologies relate to the overall health of the organism. The integrity of the intestinal epithelium is essential for the wellbeing of the organism throughout life. In recent years, intestinal barrier dysfunction has emerged as an evolutionarily conserved feature of aged organisms, as reported in worms, flies, fish, rodents and primates. Moreover, age-onset intestinal barrier dysfunction has been linked to microbial alterations, elevated immune responses, metabolic alterations, systemic health decline and mortality. Here, we provide an overview of these findings. We discuss early work in the Drosophila model that sets the stage for examining the relationship between intestinal barrier integrity and systemic aging, then delve into research in other organisms. An emerging concept, supported by studies in both Drosophila and mice, is that directly targeting intestinal barrier integrity is sufficient to promote longevity. A better understanding of the causes and consequences of age-onset intestinal barrier dysfunction has significant relevance to the development of interventions to promote healthy aging.
Collapse
Affiliation(s)
- Anna M. Salazar
- Department of Molecular Biology and Chemistry, Christopher Newport University, Newport News, VA 23606, USA
| | - Ricardo Aparicio
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095, USA
| | - Rebecca I. Clark
- Department of Biosciences, Durham University, Durham DH1 3LE, UK
| | - Michael Rera
- Université de Paris, Inserm U1284, Center for Research and Interdisciplinarity, Paris 75004, France
| | - David W. Walker
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
15
|
Yoshikawa T, Minaga K, Hara A, Sekai I, Kurimoto M, Masuta Y, Otsuka Y, Takada R, Kamata K, Park AM, Takamura S, Kudo M, Watanabe T. Disruption of the intestinal barrier exacerbates experimental autoimmune pancreatitis by promoting the translocation of Staphylococcus sciuri into the pancreas. Int Immunol 2022; 34:621-634. [PMID: 36044992 DOI: 10.1093/intimm/dxac039] [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: 02/15/2022] [Accepted: 07/27/2022] [Indexed: 02/01/2023] Open
Abstract
Autoimmune pancreatitis (AIP) and IgG4-related disease (IgG4-RD) are new disease entities characterized by enhanced IgG4 antibody responses and involvement of multiple organs, including the pancreas and salivary glands. Although the immunopathogenesis of AIP and IgG4-RD is poorly understood, we previously reported that intestinal dysbiosis mediates experimental AIP through the activation of IFN-α- and IL-33-producing plasmacytoid dendritic cells (pDCs). Because intestinal dysbiosis is linked to intestinal barrier dysfunction, we explored whether the latter affects the development of AIP and autoimmune sialadenitis in MRL/MpJ mice treated with repeated injections of polyinosinic-polycytidylic acid [poly (I:C)]. Epithelial barrier disruption was induced by the administration of dextran sodium sulfate (DSS) in the drinking water. Mice co-treated with poly (I:C) and DSS, but not those treated with either agent alone, developed severe AIP, but not autoimmune sialadenitis, which was accompanied by the increased accumulation of IFN-α- and IL-33-producing pDCs. Sequencing of 16S ribosomal RNA revealed that Staphylococcus sciuri translocation from the gut to the pancreas was preferentially observed in mice with severe AIP co-treated with DSS and poly (I:C). The degree of experimental AIP, but not of autoimmune sialadenitis, was greater in germ-free mice mono-colonized with S. sciuri and treated with poly (I:C) than in germ-free mice treated with poly (I:C) alone, which was accompanied by the increased accumulation of IFN-α- and IL-33-producing pDCs. Taken together, these data suggest that intestinal barrier dysfunction exacerbates AIP through the activation of pDCs and translocation of S. sciuri into the pancreas.
Collapse
Affiliation(s)
- Tomoe Yoshikawa
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan
| | - Kosuke Minaga
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan
| | - Akane Hara
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan
| | - Ikue Sekai
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan
| | - Masayuki Kurimoto
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan
| | - Yasuhiro Masuta
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan
| | - Yasuo Otsuka
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan
| | - Ryutaro Takada
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan
| | - Ken Kamata
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan
| | - Ah-Mee Park
- Department of Microbiology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan
| | - Shiki Takamura
- Department of Immunology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan
| | - Tomohiro Watanabe
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan
| |
Collapse
|
16
|
Liu MA, Shahabi S, Jati S, Tang K, Gao H, Jin Z, Miller W, Meunier FA, Ying W, van den Bogaart G, Ghosh G, Mahata SK. Gut microbial DNA and immune checkpoint gene Vsig4/CRIg are key antagonistic players in healthy aging and age-associated development of hypertension and diabetes. Front Endocrinol (Lausanne) 2022; 13:1037465. [PMID: 36440192 PMCID: PMC9691654 DOI: 10.3389/fendo.2022.1037465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/26/2022] [Indexed: 11/12/2022] Open
Abstract
Aims Aging is associated with the development of insulin resistance and hypertension which may stem from inflammation induced by accumulation of toxic bacterial DNA crossing the gut barrier. The aim of this study was to identify factors counter-regulating these processes. Taking advantage of the Chromogranin A (CgA) knockout (CgA-KO) mouse as a model for healthy aging, we have identified Vsig4 (V-set and immunoglobulin domain containing 4) as the critical checkpoint gene in offsetting age-associated hypertension and diabetes. Methods and Results The CgA-KO mice display two opposite aging phenotypes: hypertension but heightened insulin sensitivity at young age, whereas the blood pressure normalizes at older age and insulin sensitivity further improves. In comparison, aging WT mice gradually lost glucose tolerance and insulin sensitivity and developed hypertension. The gut barrier, compromised in aging WT mice, was preserved in CgA KO mice leading to major 35-fold protection against bacterial DNA-induced inflammation. Similarly, RNA sequencing showed increased expression of the Vsig4 gene (which removes bacterial DNA) in the liver of 2-yr-old CgA-KO mice, which may account for the very low accumulation of microbial DNA in the heart. The reversal of hypertension in aging CgA-KO mice likely stems from (i) low accumulation of microbial DNA, (ii) decreased spillover of norepinephrine in the heart and kidneys, and (iii) reduced inflammation. Conclusion We conclude that healthy aging relies on protection from bacterial DNA and the consequent low inflammation afforded by CgA-KO. Vsig4 also plays a crucial role in "healthy aging" by counteracting age-associated insulin resistance and hypertension.
Collapse
Affiliation(s)
- Matthew A. Liu
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Shandy Shahabi
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, United States
| | - Suborno Jati
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, United States
| | - Kechun Tang
- Veterans Affairs (VA) San Diego Healthcare System, San Diego, CA, United States
| | - Hong Gao
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Zhongmou Jin
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Wyatt Miller
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, United States
| | - Frédéric A. Meunier
- Clem Jones Center for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Wei Ying
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Geert van den Bogaart
- Department of Molecular Immunology and Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, Netherlands
| | - Gourisankar Ghosh
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, United States
| | - Sushil K. Mahata
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
- Veterans Affairs (VA) San Diego Healthcare System, San Diego, CA, United States
| |
Collapse
|
17
|
Giacconi R, D’Aquila P, Malavolta M, Piacenza F, Bürkle A, Villanueva MM, Dollé MET, Jansen E, Grune T, Gonos ES, Franceschi C, Capri M, Gradinaru D, Grubeck-Loebenstein B, Sikora E, Stuetz W, Weber D, Toussaint O, Debacq-Chainiaux F, Hervonen A, Hurme M, Slagboom PE, Schön C, Bernhardt J, Breusing N, Duncan T, Passarino G, Bellizzi D, Provinciali M. Bacterial DNAemia in Older Participants and Nonagenarian Offspring and Association With Redox Biomarkers: Results From MARK-AGE Study. J Gerontol A Biol Sci Med Sci 2022; 78:42-50. [PMID: 35914804 PMCID: PMC9879758 DOI: 10.1093/gerona/glac154] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Indexed: 02/02/2023] Open
Abstract
Aging and age-related diseases have been linked to microbial dysbiosis with changes in blood bacterial DNA concentration. This condition may promote chronic low-grade inflammation, which can be further aggravated by antioxidant nutrient deficiency. Low plasma carotenoids are associated with an increased risk of inflammation and cellular damage and predict mortality. However, no evidence is yet available on the relationship between antioxidants and the blood bacterial DNA (BB-DNA). Therefore, this study aimed to compare BB-DNA from (a) GO (nonagenarian offspring), (b) age-matched controls (Randomly recruited Age-Stratified Individuals from the General population [RASIG]), and (c) spouses of GO (SGO) recruited in the MARK-AGE project, as well as to investigate the association between BB-DNA, behavior habits, Charlson Comorbidity Index (CCI), leucocyte subsets, and the circulating levels of some antioxidants and oxidative stress markers. BB-DNA was higher in RASIG than GO and SGO, whereas GO and SGO participants showed similar values. BB-DNA increased in smokers and males with CCI ≥ 2 compared with those with CCI ≤ 1 within RASIG. Moreover, BB-DNA was positively associated with lymphocyte, neutrophil, and monocyte counts, but not with self-reported dietary habits. Higher quartiles of BB-DNA were associated with low lutein and zeaxanthin and elevated malondialdehyde plasma concentrations in RASIG. BB-DNA was also positively correlated with nitric oxide levels. Herein, we provide evidence of a reduced BB-DNA in individuals from long-living families and their spouses, suggesting a decreased microbial dysbiosis and bacterial systemic translocation. BB-DNA was also associated with smoking, CCI, leukocyte subsets, and some redox biomarkers in older participants.
Collapse
Affiliation(s)
- Robertina Giacconi
- Address correspondence to: Robertina Giacconi, Advanced Technology Center for Aging Research, IRCCS INRCA, via birarelli 8 Ancona, 60121 Ancona, Italy. E-mail:
| | | | - Marco Malavolta
- Advanced Technology Center for Aging Research, IRCCS INRCA, Ancona, Italy
| | - Francesco Piacenza
- Advanced Technology Center for Aging Research, IRCCS INRCA, Ancona, Italy
| | - Alexander Bürkle
- Molecular Toxicology Group, Department of Biology, University of Konstanz, Konstanz, Germany
| | - María Moreno Villanueva
- Molecular Toxicology Group, Department of Biology, University of Konstanz, Konstanz, Germany,Human Performance Research Centre, Department of Sport Science, University of Konstanz, Konstanz, Germany
| | - Martijn E T Dollé
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Eugène Jansen
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Tilman Grune
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany,University of Potsdam, Institute of Nutritional Science, Nuthetal, Germany,Department of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Efstathios S Gonos
- National Hellenic Research Foundation, Institute of Biology, Medicinal Chemistry and Biotechnology, Athens, Greece
| | - Claudio Franceschi
- Department of Experimental, Diagnostic and Specialty Medicine, Alma Mater Studiorum, University of Bologna, Bologna, Italy,Institute of Information Technologies, Mathematics and Mechanics, Lobachevsky University, Nizhniy Novgorod, Russia
| | - Miriam Capri
- Department of Experimental, Diagnostic and Specialty Medicine, Alma Mater Studiorum, University of Bologna, Bologna, Italy,Interdepartmental Center—Alma Mater Research Institute on Global Challenges and Climate Change, University of Bologna, Bologna, Italy
| | - Daniela Gradinaru
- Ana Aslan National Institute of Gerontology and Geriatrics, Bucharest, Romania,Faculty of Pharmacy, Department of Biochemistry, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | | | - Ewa Sikora
- Laboratory of the Molecular Bases of Ageing, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Wolfgang Stuetz
- Institute of Nutritional Sciences, Department of Food Biofunctionality, University of Hohenheim, Stuttgart, Germany
| | - Daniela Weber
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany,University of Potsdam, Institute of Nutritional Science, Nuthetal, Germany
| | | | | | - Antti Hervonen
- The Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Mikko Hurme
- The Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - P Eline Slagboom
- Department of Molecular Epidemiology, Leiden University Medical Centre, Leiden, The Netherlands
| | | | | | - Nicolle Breusing
- Department of Applied Nutritional Science/Dietetics, Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | | | - Giuseppe Passarino
- Department of Biology, Ecology and Earth Sciences (DIBEST), University of Calabria, Rende, Italy
| | - Dina Bellizzi
- Department of Biology, Ecology and Earth Sciences (DIBEST), University of Calabria, Rende, Italy
| | | |
Collapse
|
18
|
Milenkovic D, Capel F, Combaret L, Comte B, Dardevet D, Evrard B, Guillet C, Monfoulet LE, Pinel A, Polakof S, Pujos-Guillot E, Rémond D, Wittrant Y, Savary-Auzeloux I. Targeting the gut to prevent and counteract metabolic disorders and pathologies during aging. Crit Rev Food Sci Nutr 2022; 63:11185-11210. [PMID: 35730212 DOI: 10.1080/10408398.2022.2089870] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Impairment of gut function is one of the explanatory mechanisms of health status decline in elderly population. These impairments involve a decline in gut digestive physiology, metabolism and immune status, and associated to that, changes in composition and function of the microbiota it harbors. Continuous deteriorations are generally associated with the development of systemic dysregulations and ultimately pathologies that can worsen the initial health status of individuals. All these alterations observed at the gut level can then constitute a wide range of potential targets for development of nutritional strategies that can impact gut tissue or associated microbiota pattern. This can be key, in a preventive manner, to limit gut functionality decline, or in a curative way to help maintaining optimum nutrients bioavailability in a context on increased requirements, as frequently observed in pathological situations. The aim of this review is to give an overview on the alterations that can occur in the gut during aging and lead to the development of altered function in other tissues and organs, ultimately leading to the development of pathologies. Subsequently is discussed how nutritional strategies that target gut tissue and gut microbiota can help to avoid or delay the occurrence of aging-related pathologies.
Collapse
Affiliation(s)
- Dragan Milenkovic
- Human Nutrition Unit, UMR1019, University Clermont Auvergne, INRAE, Clermont-Ferrand, France
| | - Frédéric Capel
- Human Nutrition Unit, UMR1019, University Clermont Auvergne, INRAE, Clermont-Ferrand, France
| | - Lydie Combaret
- Human Nutrition Unit, UMR1019, University Clermont Auvergne, INRAE, Clermont-Ferrand, France
| | - Blandine Comte
- Human Nutrition Unit, UMR1019, University Clermont Auvergne, INRAE, Clermont-Ferrand, France
| | - Dominique Dardevet
- Human Nutrition Unit, UMR1019, University Clermont Auvergne, INRAE, Clermont-Ferrand, France
| | - Bertrand Evrard
- Human Nutrition Unit, UMR1019, University Clermont Auvergne, INRAE, Clermont-Ferrand, France
| | - Christelle Guillet
- Human Nutrition Unit, UMR1019, University Clermont Auvergne, INRAE, Clermont-Ferrand, France
| | | | - Alexandre Pinel
- Human Nutrition Unit, UMR1019, University Clermont Auvergne, INRAE, Clermont-Ferrand, France
| | - Sergio Polakof
- Human Nutrition Unit, UMR1019, University Clermont Auvergne, INRAE, Clermont-Ferrand, France
| | - Estelle Pujos-Guillot
- Human Nutrition Unit, UMR1019, University Clermont Auvergne, INRAE, Clermont-Ferrand, France
| | - Didier Rémond
- Human Nutrition Unit, UMR1019, University Clermont Auvergne, INRAE, Clermont-Ferrand, France
| | - Yohann Wittrant
- Human Nutrition Unit, UMR1019, University Clermont Auvergne, INRAE, Clermont-Ferrand, France
| | | |
Collapse
|
19
|
Kadyan S, Sharma A, Arjmandi BH, Singh P, Nagpal R. Prebiotic Potential of Dietary Beans and Pulses and Their Resistant Starch for Aging-Associated Gut and Metabolic Health. Nutrients 2022; 14:nu14091726. [PMID: 35565693 PMCID: PMC9100130 DOI: 10.3390/nu14091726] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/18/2022] [Accepted: 04/19/2022] [Indexed: 01/27/2023] Open
Abstract
Dietary pulses, including dry beans, lentils, chickpeas, and dry peas, have the highest proportion of fiber among different legume cultivars and are inexpensive, easily accessible, and have a long shelf-life. The inclusion of pulses in regular dietary patterns is an easy and effective solution for achieving recommended fiber intake and maintaining a healthier gut and overall health. Dietary pulses-derived resistant starch (RS) is a relatively less explored prebiotic ingredient. Several in vitro and preclinical studies have elucidated the crucial role of RS in fostering and shaping the gut microbiota composition towards homeostasis thereby improving host metabolic health. However, in humans and aged animal models, the effect of only the cereals and tubers derived RS has been studied. In this context, this review collates literature pertaining to the beneficial effects of dietary pulses and their RS on gut microbiome-metabolome signatures in preclinical and clinical studies while contemplating their potential and prospects for better aging-associated gut health. In a nutshell, the incorporation of dietary pulses and their RS in diet fosters the growth of beneficial gut bacteria and significantly enhances the production of short-chain fatty acids in the colon.
Collapse
|
20
|
Kramer CS, Groenendijk I, Beers S, Wijnen HH, van de Rest O, de Groot LCPGM. The Association between Malnutrition and Physical Performance in Older Adults: A Systematic Review and Meta-Analysis of Observational Studies. Curr Dev Nutr 2022; 6:nzac007. [PMID: 35415390 PMCID: PMC8989279 DOI: 10.1093/cdn/nzac007] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/14/2021] [Indexed: 11/13/2022] Open
Abstract
In recent years the focus of healthcare and nutritional science in older adults has shifted from mortality towards physical performance and quality of life. The aim of this review was to summarize observational studies on physical performance in malnourished (MN) or at risk of malnutrition (RMN) older adults compared with well-nourished (WN) older adults. Eligible studies had to report on nutritional status and objectively measured physical performance in older adults (≥60 y). MN or RMN groups had to be compared with a WN group, measured with a validated nutrition screener. Ovid Medline and Web of Science were searched until 13 November, 2020. Study quality was scored using a modified Newcastle-Ottawa Scale (NOS). Results were analyzed by meta-analysis when possible, or narratively reviewed otherwise. Forty-five studies (16,911 participants in total) were included from studies in outpatient clinics (n = 6), nursing homes (n = 3), community-dwelling older adults (n = 20), hospitalized patients (n = 15), or a combination (n = 1). Studies used 11 different screeners of malnutrition, and 8 types of physical performance measures. Meta-analysis showed that compared with MN, WN groups had better hand grip strength (mean difference [MD] = 4.92 kg; 95% CI: 3.43, 6.41; P < 0.001; n = 23), faster gait speed (MD = 0.16 m/s; 95% CI: 0.05, 0.27; P = 0.0033; n = 7), performed faster on timed-up-and-go (MD = -5.94 s; 95% CI: -8.98, -2.89; P < 0.001; n = 8), and scored 1.2 more short physical performance battery points (95% CI: 1.32, 2.73; P < 0.001; n = 6). Results were less pronounced when compared with RMN. Narratively, all studies showed an association for knee extension strength, 6-min walking test, and multicomponent tests, except for the chair stand test. Study limitations include no studies scoring "good" on NOS, lack of confounder adjustment, and high heterogeneity. Overall, evidence from cross-sectional studies indicate an association between malnutrition and worse physical performance in older adults. This study is registered in PROSPERO as CRD42020192893.
Collapse
Affiliation(s)
- Charlotte S Kramer
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Inge Groenendijk
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Sonja Beers
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Hugo H Wijnen
- Department of Geriatrics, Rijnstate Hospital, Arnhem, The Netherlands
| | - Ondine van de Rest
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Lisette C P G M de Groot
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| |
Collapse
|
21
|
Vemuri R, Ruggiero A, Whitfield JM, Dugan GO, Cline JM, Block MR, Guo H, Kavanagh K. Hypertension promotes microbial translocation and dysbiotic shifts in the fecal microbiome of nonhuman primates. Am J Physiol Heart Circ Physiol 2022; 322:H474-H485. [PMID: 35148233 PMCID: PMC8897002 DOI: 10.1152/ajpheart.00530.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Accumulating evidence indicates a link between gut barrier dysfunction and hypertension. However, it is unclear whether hypertension causes gut barrier dysfunction or vice versa and whether the gut microbiome plays a role. To understand this relationship, we first cross-sectionally examined 153 nonhuman primates [NHPs; Chlorocebus aethiops sabaeus; mean age, 16 ± 0.4 yr; 129 (84.3%) females] for cardiometabolic risk factors and gut barrier function biomarkers. This analysis identified blood pressure and age as specific factors that independently associated with microbial translocation. We then longitudinally tracked male, age-matched spontaneously hypertensive NHPs (Macaca mulatta) to normotensives (n = 16), mean age of 5.8 ± 0.5 yr, to confirm hypertension-related gut barrier dysfunction and to explore the role of microbiome by comparing groups at baseline, 12, and 27 mo. Collectively, hypertensive animals in both studies showed evidence of gut barrier dysfunction (i.e., microbial translocation), as indicated by higher plasma levels of lipopolysaccharide-binding protein (LBP)-1, when compared with normotensive animals. Furthermore, plasma LBP-1 levels were correlated with diastolic blood pressure, independent of age and other health markers, suggesting specificity of the effect of hypertension on microbial translocation. In over 2 yr of longitudinal assessment, hypertensive animals had escalating plasma levels of LBP-1 and greater bacterial gene expression in mesenteric lymph nodes compared with normotensive animals, confirming microbes translocated across the intestinal barrier. Concomitantly, we identified distinct shifts in the gut microbial signature of hypertensive versus normotensive animals at 12 and 27 mo. These results suggest that hypertension contributes to microbial translocation in the gut and eventually unhealthy shifts in the gut microbiome, possibly contributing to poor health outcomes, providing further impetus for the management of hypertension.NEW & NOTEWORTHY Hypertension specifically had detrimental effects on microbial translocation when age and metabolic syndrome criteria were evaluated as drivers of cardiovascular disease in a relevant nonhuman primate model. Intestinal barrier function exponentially decayed over time with chronic hypertension, and microbial translocation was confirmed by detection of more microbial genes in regional draining lymph nodes. Chronic hypertension resulted in fecal microbial dysbiosis and elevations of the biomarker NT-proBNP. This study provides insights on the barrier dysfunction, dysbiosis, and hypertension in controlled studies of nonhuman primates. Our study includes a longitudinal component comparing naturally occurring hypertensive to normotensive primates to confirm microbial translocation and dysbiotic microbiome development. Hypertension is an underappreciated driver of subclinical endotoxemia that can drive chronic inflammatory diseases.
Collapse
Affiliation(s)
- Ravichandra Vemuri
- 1Section on Comparative Medicine, Department of Pathology, Wake Forest School of Medicine, Winston‐Salem, North Carolina
| | - Alistaire Ruggiero
- 1Section on Comparative Medicine, Department of Pathology, Wake Forest School of Medicine, Winston‐Salem, North Carolina
| | - Jordyn M. Whitfield
- 1Section on Comparative Medicine, Department of Pathology, Wake Forest School of Medicine, Winston‐Salem, North Carolina
| | - Greg O. Dugan
- 1Section on Comparative Medicine, Department of Pathology, Wake Forest School of Medicine, Winston‐Salem, North Carolina
| | - J. Mark Cline
- 1Section on Comparative Medicine, Department of Pathology, Wake Forest School of Medicine, Winston‐Salem, North Carolina
| | - Masha R. Block
- 1Section on Comparative Medicine, Department of Pathology, Wake Forest School of Medicine, Winston‐Salem, North Carolina
| | - Hao Guo
- 2Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina,3Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Kylie Kavanagh
- 1Section on Comparative Medicine, Department of Pathology, Wake Forest School of Medicine, Winston‐Salem, North Carolina,4Department of Biomedicine, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
22
|
Vemuri R, Martoni CJ, Kavanagh K, Eri R. Lactobacillus acidophilus DDS-1 Modulates the Gut Microbial Co-Occurrence Networks in Aging Mice. Nutrients 2022; 14:nu14050977. [PMID: 35267950 PMCID: PMC8912519 DOI: 10.3390/nu14050977] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 02/18/2022] [Accepted: 02/23/2022] [Indexed: 12/13/2022] Open
Abstract
Age-related alterations in the gut microbiome composition and its impacts on the host’s health have been well-described; however, detailed analyses of the gut microbial structure defining ecological microbe–microbe interactions are limited. One of the ways to determine these interactions is by understanding microbial co-occurrence patterns. We previously showed promising abilities of Lactobacillus acidophilus DDS-1 on the aging gut microbiome and immune system. However, the potential of the DDS-1 strain to modulate microbial co-occurrence patterns is unknown. Hence, we aimed to investigate the ability of L. acidophilus DDS-1 to modulate the fecal-, mucosal-, and cecal-related microbial co-occurrence networks in young and aging C57BL/6J mice. Our Kendall’s tau correlation measures of co-occurrence revealed age-related changes in the gut microbiome, which were characterized by a reduced number of nodes and associations across sample types when compared to younger mice. After four-week supplementation, L. acidophilus DDS-1 differentially modulated the overall microbial community structure in fecal and mucosal samples as compared to cecal samples. Beneficial bacteria such as Lactobacillus, Oscillospira, and Akkermansia acted as connectors in aging networks in response to L. acidophilus DDS-1 supplementation. Our findings provided the first evidence of the DDS-1-induced gut microbial ecological interactions, revealing the complex structure of microbial ecosystems with age.
Collapse
Affiliation(s)
- Ravichandra Vemuri
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA;
- College of Health and Medicine, School of Health Sciences, University of Tasmania, Launceston, TAS 7248, Australia;
- Correspondence:
| | | | - Kylie Kavanagh
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA;
- Department of Biomedicine, University of Tasmania, Hobart, TAS 7000, Australia
| | - Rajaraman Eri
- College of Health and Medicine, School of Health Sciences, University of Tasmania, Launceston, TAS 7248, Australia;
| |
Collapse
|
23
|
Kim E, Lee HG, Han S, Seo KH, Kim H. Effect of Surface Layer Proteins Derived from Paraprobiotic Kefir Lactic Acid Bacteria on Inflammation and High-Fat Diet-Induced Obesity. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:15157-15164. [PMID: 34882385 DOI: 10.1021/acs.jafc.1c05037] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The antiobesity action of nonviable probiotic lactic acid bacteria (PLAB) may be attributed to bacterial cellular components recognized by host cells. The anti-inflammation and antiobesity properties of surface layer proteins (SLPs) that are cellular components isolated from kefir PLAB were determined in macrophage RAW 264.7 cells and obese mice. Kefir SLPs significantly decreased secretion of IL-6 and production of NF-kB p65 protein by LPS-stimulated RAW 264.7 cells in a dose-response manner. C57BL/6J mice were fed a high-fat (HF) diet with oral administration of either saline (CON) or kefir SLPs for 6 weeks. SLPs significantly improved body weight gain and adipose tissue weight, plasma triglyceride concentrations, and insulin resistance. Profiling of adipocyte gene expression showed that the antiobesity effect was significantly related to the expression of genes associated with adipogenesis, autophagy, and inflammatory/immune response, and fatty acid oxidation. Taken together, SLPs are a novel bioactive component in kefir PLABs to target obesity and obesity-related disorders.
Collapse
Affiliation(s)
- Eseul Kim
- Department of Food and Nutrition, Hanyang University, Seoul 04763, South Korea
| | - Hyeon Gyu Lee
- Department of Food and Nutrition, Hanyang University, Seoul 04763, South Korea
| | - Sanghoon Han
- Department of Food and Nutrition, Hanyang University, Seoul 04763, South Korea
| | - Kun-Ho Seo
- Center for One Health, College of Veterinary Medicine, Konkuk University, Seoul 05029, South Korea
| | - Hyunsook Kim
- Department of Food and Nutrition, Hanyang University, Seoul 04763, South Korea
| |
Collapse
|
24
|
Wang L, Zhang P, Li C, Xu F, Chen J. A polysaccharide from Rosa roxburghii Tratt fruit attenuates high-fat diet-induced intestinal barrier dysfunction and inflammation in mice by modulating the gut microbiota. Food Funct 2021; 13:530-547. [PMID: 34932054 DOI: 10.1039/d1fo03190b] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Obesity-induced colonic inflammation-stimulated colitis is one of the main causes of colorectal cancer. Dietary polysaccharides are considered an effective agent for relieving obesity-induced inflammatory diseases such as diabetes and colitis. In this work, the protective effects of a polysaccharide (RTFP) extracted from Rosa roxburghii Tratt fruit on barrier dysfunction and inflammation were investigated using obesity-induced colitis model mice. RTFP treatment repaired intestinal barrier dysfunction by increasing the expression of tight junction proteins (ZO-1, claudin-1, and occludin) and reducing the levels of inflammatory cytokines, intestinal permeability, and colonic oxidative stress in mice fed a high-fat diet. Most significantly, RTFP decreased gut inflammation and ameliorated the metabolic dysbiosis of intestinal microflora by decreasing the Firmicutes/Bacteroidetes ratio, reducing the levels of serum D-lactic acid and lipopolysaccharides, and inhibiting the TLR4/NF-κB signaling pathway. Furthermore, RTFP significantly increased the abundance of beneficial bacteria (Ruminococcaceae, Muribaculaceae, Akkermansiaceae, etc.) but decreased the abundance of pathogenic bacteria. These findings indicate that RTFP can be used as a natural anti-inflammatory agent to reduce chronic obesity-induced colitis.
Collapse
Affiliation(s)
- Lei Wang
- College of Food Science and Engineering, Henan University of Technology, Zhengzhou, 450001, China.
| | - Pan Zhang
- College of Food Science and Engineering, Henan University of Technology, Zhengzhou, 450001, China.
| | - Chao Li
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Fei Xu
- College of Food Science and Engineering, Henan University of Technology, Zhengzhou, 450001, China.
| | - Jie Chen
- College of Food Science and Engineering, Henan University of Technology, Zhengzhou, 450001, China.
| |
Collapse
|
25
|
Frye BM, Craft S, Latimer CS, Keene CD, Montine TJ, Register TC, Orr ME, Kavanagh K, Macauley SL, Shively CA. Aging-related Alzheimer's disease-like neuropathology and functional decline in captive vervet monkeys (Chlorocebus aethiops sabaeus). Am J Primatol 2021; 83:e23260. [PMID: 33818801 PMCID: PMC8626867 DOI: 10.1002/ajp.23260] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 03/05/2021] [Accepted: 03/21/2021] [Indexed: 12/17/2022]
Abstract
Age-related neurodegeneration characteristic of late-onset Alzheimer's disease (LOAD) begins in middle age, well before symptoms. Translational models to identify modifiable risk factors are needed to understand etiology and identify therapeutic targets. Here, we outline the evidence supporting the vervet monkey (Chlorocebus aethiops sabaeus) as a model of aging-related AD-like neuropathology and associated phenotypes including cognitive function, physical function, glucose handling, intestinal physiology, and CSF, blood, and neuroimaging biomarkers. This review provides the most comprehensive multisystem description of aging in vervets to date. This review synthesizes a large body of evidence that suggests that aging vervets exhibit a coordinated suite of traits consistent with early AD and provide a powerful, naturally occurring model for LOAD. Notably, relationships are identified between AD-like neuropathology and modifiable risk factors. Gaps in knowledge and key limitations are provided to shape future studies to illuminate mechanisms underlying divergent neurocognitive aging trajectories and to develop interventions that increase resilience to aging-associated chronic disease, particularly, LOAD.
Collapse
Affiliation(s)
- Brett M. Frye
- Department of Pathology/Comparative Medicine, Wake Forest School of Medicine
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine
| | - Suzanne Craft
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine
- Wake Forest Alzheimer’s Disease Research Center
- J. Paul Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest School of Medicine
| | - Caitlin S. Latimer
- Department of Laboratory Medicine and Pathology, University of Washington-Seattle
| | - C. Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington-Seattle
| | | | - Thomas C. Register
- Department of Pathology/Comparative Medicine, Wake Forest School of Medicine
- Wake Forest Alzheimer’s Disease Research Center
- J. Paul Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest School of Medicine
| | - Miranda E. Orr
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine
| | - Kylie Kavanagh
- Department of Pathology/Comparative Medicine, Wake Forest School of Medicine
| | - Shannon L. Macauley
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine
- Wake Forest Alzheimer’s Disease Research Center
| | - Carol A. Shively
- Department of Pathology/Comparative Medicine, Wake Forest School of Medicine
- Wake Forest Alzheimer’s Disease Research Center
| |
Collapse
|
26
|
Huang YJ, Huang CJ. Construction of a 5 immune-related lncRNA-based prognostic model of NSCLC via bioinformatics. Medicine (Baltimore) 2021; 100:e27222. [PMID: 34664861 PMCID: PMC8448051 DOI: 10.1097/md.0000000000027222] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 08/28/2021] [Indexed: 11/25/2022] Open
Abstract
Participate in tumorigenic, oncogenic, and tumor suppressive pathways through gene expression regulation. We aimed to build an immune-related long noncoding RNA (lncRNA) prognostic model to enhance nonsmall cell lung cancer (NSCLC) prognostic prediction.The original data were collected from the cancer genome atlas database. Perl and R software were used for statistical analysis. The effects of lncRNAs expression on prognosis were analyzed by Gene Expression Profiling Interactive Analysis. Silico functional analysis were performed by DAVID Bioinformatics Resources.The median risk score as a dividing value separated patients into high- and low-risk groups. These 2 groups had different 5-year survival rates, median survival times, and immune statuses. The 5-lncRNA signature was validated as an independent prognostic factor with high accuracy (area under the receiver operating characteristic = 0.722). Silico functional analysis connected the lncRNAs with immune-related biological processes and pathways in carcinogenesis.The novel immune-related lncRNA prognostic model had significant clinical implication for enhancing lung adenocarcinoma outcome prediction and guiding the choice of treatment.
Collapse
Affiliation(s)
- Ya-jie Huang
- Department of Medical Oncology, The Fourth hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Chang-jie Huang
- Undergraduate of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
27
|
Ogita T, Namai F, Mikami A, Ishiguro T, Umezawa K, Uyeno Y, Shimosato T. A Soybean Resistant Protein-Containing Diet Increased the Production of Reg3γ Through the Regulation of the Gut Microbiota and Enhanced the Intestinal Barrier Function in Mice. Front Nutr 2021; 8:701466. [PMID: 34490323 PMCID: PMC8416681 DOI: 10.3389/fnut.2021.701466] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 07/27/2021] [Indexed: 12/12/2022] Open
Abstract
The maintenance of intestinal homeostasis is necessary for a good quality of life, and strengthening of the intestinal barrier function is thus an important issue. Therefore, we focused on soybean resistant protein (SRP) derived from kori-tofu (freeze-dried tofu), which is a traditional Japanese food, as a functional food component. In this study, to investigate the effect of SRP on the intestinal barrier function and intestinal microbiota, we conducted an SRP free intake experiment in mice. Results showed that ingestion of SRP decreased the serum level of lipopolysaccharide-binding protein and induced the expression of Reg3γ, thereby improving the intestinal barrier function. In addition, SRP intake induced changes in the cecal microbiota, as observed by changes in β-diversity. In particular, in the microbiota, the up-regulation of functional gene pathways related to the bacterial invasion of epithelial cells (ko05100) was observed, suggesting that Reg3γ expression was induced by the direct stimulation of epithelial cells. The results of this study suggest that SRP is a functional food component that may contribute to the maintenance of intestinal homeostasis.
Collapse
Affiliation(s)
- Tasuku Ogita
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| | - Fu Namai
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan.,Department of Pathology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Ayane Mikami
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| | | | - Koji Umezawa
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| | - Yutaka Uyeno
- Faculty of Agriculture, Shinshu University, Nagano, Japan
| | - Takeshi Shimosato
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| |
Collapse
|
28
|
de Marco Castro E, Murphy CH, Roche HM. Targeting the Gut Microbiota to Improve Dietary Protein Efficacy to Mitigate Sarcopenia. Front Nutr 2021; 8:656730. [PMID: 34235167 PMCID: PMC8256992 DOI: 10.3389/fnut.2021.656730] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 05/24/2021] [Indexed: 12/11/2022] Open
Abstract
Sarcopenia is characterised by the presence of diminished skeletal muscle mass and strength. It is relatively common in older adults as ageing is associated with anabolic resistance (a blunted muscle protein synthesis response to dietary protein consumption and resistance exercise). Therefore, interventions to counteract anabolic resistance may benefit sarcopenia prevention and are of utmost importance in the present ageing population. There is growing speculation that the gut microbiota may contribute to sarcopenia, as ageing is also associated with [1) dysbiosis, whereby the gut microbiota becomes less diverse, lacking in healthy butyrate-producing microorganisms and higher in pathogenic bacteria, and [2) loss of epithelial tight junction integrity in the lining of the gut, leading to increased gut permeability and higher metabolic endotoxemia. Animal data suggest that both elements may impact muscle physiology, but human data corroborating the causality of the association between gut microbiota and muscle mass and strength are lacking. Mechanisms wherein the gut microbiota may alter anabolic resistance include an attenuation of gut-derived low-grade inflammation and/or the increased digestibility of protein-containing foods and consequent higher aminoacidemia, both in favour of muscle protein synthesis. This review focuses on the putative links between the gut microbiota and skeletal muscle in the context of sarcopenia. We also address the issue of plant protein digestibility because plant proteins are increasingly important from an environmental sustainability perspective, yet they are less efficient at stimulating muscle protein synthesis than animal proteins.
Collapse
Affiliation(s)
- Elena de Marco Castro
- Nutrigenomics Research Group, School of Public Health, Physiotherapy, and Sports Science, UCD Conway Institute, UCD Institute of Food and Health, University College Dublin, Dublin, Ireland
| | - Caoileann H Murphy
- Nutrigenomics Research Group, School of Public Health, Physiotherapy, and Sports Science, UCD Conway Institute, UCD Institute of Food and Health, University College Dublin, Dublin, Ireland.,Teagasc Food Research Centre, Ashtown, Dublin, Ireland
| | - Helen M Roche
- Nutrigenomics Research Group, School of Public Health, Physiotherapy, and Sports Science, UCD Conway Institute, UCD Institute of Food and Health, University College Dublin, Dublin, Ireland.,Institute for Global Food Security, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
29
|
Proshkina EN, Solovev IA, Shaposhnikov MV, Moskalev AA. Key Molecular Mechanisms of Aging, Biomarkers, and Potential Interventions. Mol Biol 2021. [DOI: 10.1134/s0026893320060096] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
30
|
Introduction to host microbiome symbiosis in health and disease. Mucosal Immunol 2021; 14:547-554. [PMID: 33299088 PMCID: PMC7724625 DOI: 10.1038/s41385-020-00365-4] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 02/06/2023]
Abstract
Humans share a core intestinal microbiome and yet human microbiome differs by genes, species, enterotypes (ecology), and gene count (microbial diversity). Achievement of microbiota metagenomic analysis has revealed that the microbiome gene count is a key stratifier of health in several immune disorders and clinical conditions. We review here the progress of the metagenomic pipeline analysis, and how this has allowed us to define the host-microbe symbiosis associated with a healthy status. The link between host-microbe symbiosis disruption, the so-called dysbiosis and chronic diseases or iatrogenic conditions is highlighted. Finally, opportunities to use microbiota modulation, with specific nutrients and/or live microbes, as a target for personalized nutrition and therapy for the maintenance, preservation, or restoration of host-microbe symbiosis are discussed.
Collapse
|
31
|
Specificity Influences in (1→3)-β-d-Glucan-Supported Diagnosis of Invasive Fungal Disease. J Fungi (Basel) 2020; 7:jof7010014. [PMID: 33383818 PMCID: PMC7824349 DOI: 10.3390/jof7010014] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/17/2020] [Accepted: 12/21/2020] [Indexed: 12/12/2022] Open
Abstract
(1→3)-β-glucan (BDG) testing as an adjunct in the diagnosis of invasive fungal disease (IFD) has been in use for nearly three decades. While BDG has a very high negative predictive value in this setting, diagnostic false positives may occur, limiting specificity and positive predictive value. Although results may be diagnostically false positive, they are analytically correct, due to the presence of BDG in the circulation. This review surveys the non-IFD causes of elevated circulating BDG. These are in the main, iatrogenic patient contamination through the use of BDG-containing medical devices and parenterally-delivered materials as well as translocation of intestinal luminal BDG due to mucosal barrier injury. Additionally, infection with Nocardia sp. may also contribute to elevated circulating BDG. Knowledge of the factors which may contribute to such non-IFD-related test results can improve the planning and interpretation of BDG assays and permit investigational strategies, such as serial sampling and BDG clearance evaluation, to assess the likelihood of contamination and improve patient care.
Collapse
|
32
|
Bhattacharya S, Bhadra R, Schols AMWJ, Sambashivaiah S. Gut microbial dysbiosis as a limiting factor in the management of primary and secondary sarcopenia: an Asian Indian perspective. Curr Opin Clin Nutr Metab Care 2020; 23:404-410. [PMID: 32868683 DOI: 10.1097/mco.0000000000000688] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE OF REVIEW The article summarizes recent research advances on the role of gut microbiome in primary and secondary sarcopenia. This article also explores the potential contribution of gut dysbiosis to suboptimal sarcopenia management with special focus on factors contributing to gut dysbiosis among Asian Indians. RECENT FINDINGS Aging and chronic diseases contribute to gut dysbiosis and intestinal barrier dysfunction allowing enhanced microbial translocation that may negatively affect muscle strength, physical function, and frailty. Gut microbiome of Asian Indians has shown a unique composition that is affected by multiple factors, such as socioeconomic status, poor hygiene, high rate of infection and infestations, antibiotic overuse and transition towards a westernized eating pattern. Current management approach for sarcopenia (exercise and/or protein supplementation) fails to address gut dysbiosis and intestinal barrier dysfunction. Incorporating a prebiotic or probiotic element to the intervention strategy may improve gut dysbiosis, inflammation and muscle function. SUMMARY Gut dysbiosis and intestinal barrier dysfunction appear to be a significant limitation in sarcopenia management, thus gut centric intervention may be perceived as a (co)intervention strategy to be tested in appropriate clinical trials.
Collapse
Affiliation(s)
| | - Rohini Bhadra
- St John's Research Institute, St John's Medical College, Bengaluru, India
| | - Annemie M W J Schols
- NUTRIM School of Nutrition and Translational Research in Metabolism, Department of Respiratory Medicine, Maastricht University, The Netherlands
| | | |
Collapse
|
33
|
Sári Z, Kovács T, Csonka T, Török M, Sebő É, Toth J, Tóth D, Mikó E, Kiss B, Szeőcs D, Uray K, Karányi Z, Kovács I, Méhes G, Árkosy P, Bai P. Fecal expression of Escherichia coli lysine decarboxylase (LdcC) is downregulated in E-cadherin negative lobular breast carcinoma. Physiol Int 2020; 107:349-358. [PMID: 32692716 DOI: 10.1556/2060.2020.00016] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 05/21/2020] [Indexed: 11/19/2022]
Abstract
Breast cancer is characterized by oncobiosis, the abnormal composition of the microbiome in neoplastic diseases. The biosynthetic capacity of the oncobiotic flora in breast cancer is suppressed, as suggested by metagenomic studies. The microbiome synthesizes a set of cytostatic and antimetastatic metabolites that are downregulated in breast cancer, including cadaverine, a microbiome metabolite with cytostatic properties. We set out to assess how the protein expression of constitutive lysine decarboxylase (LdcC), a key enzyme for cadaverine production, changes in the feces of human breast cancer patients (n = 35). We found that the fecal expression of Escherichia coli LdcC is downregulated in lobular cases as compared to invasive carcinoma of no special type (NST) cases. Lobular breast carcinoma is characterized by low or absent expression of E-cadherin. Fecal E. coli LdcC protein expression is downregulated in E-cadherin negative breast cancer cases as compared to positive ones. Receiver operating characteristic (ROC) analysis of LdcC expression in lobular and NST cases revealed that fecal E. coli LdcC protein expression might have predictive values. These data suggest that the oncobiotic transformation of the microbiome indeed leads to the downregulation of the production of cytostatic and antimetastatic metabolites. In E-cadherin negative lobular carcinoma that has a higher potential for metastasis formation, the protein levels of enzymes producing antimetastatic metabolites are downregulated. This finding represents a new route that renders lobular cases permissive for metastasis formation. Furthermore, our findings underline the role of oncobiosis in regulating metastasis formation in breast cancer.
Collapse
Affiliation(s)
- Zs Sári
- 1Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - T Kovács
- 1Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - T Csonka
- 2Department of Pathology, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - M Török
- 3Department of Pathology, Kenézy Gyula County Hospital, Debrecen, 4032, Hungary
| | - É Sebő
- 4Kenézy Breast Center, Kenézy Gyula County Hospital, Debrecen, 4032, Hungary
| | - J Toth
- 5Department of Oncology, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - D Tóth
- 6Department of Surgery, Borsod-Abaúj-Zemplén County Hospital and University Teaching Hospital, Miskolc, 3526, Hungary
| | - E Mikó
- 1Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - B Kiss
- 5Department of Oncology, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - D Szeőcs
- 1Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - K Uray
- 1Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Zs Karányi
- 7Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - I Kovács
- 3Department of Pathology, Kenézy Gyula County Hospital, Debrecen, 4032, Hungary
| | - G Méhes
- 2Department of Pathology, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - P Árkosy
- 5Department of Oncology, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - P Bai
- 1Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary.,8MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, 4032, Hungary.,9Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| |
Collapse
|
34
|
Ahmadi S, Wang S, Nagpal R, Wang B, Jain S, Razazan A, Mishra SP, Zhu X, Wang Z, Kavanagh K, Yadav H. A human-origin probiotic cocktail ameliorates aging-related leaky gut and inflammation via modulating the microbiota/taurine/tight junction axis. JCI Insight 2020; 5:132055. [PMID: 32302292 DOI: 10.1172/jci.insight.132055] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 04/08/2020] [Indexed: 12/25/2022] Open
Abstract
Inflammation is a major risk factor of morbidity and mortality in older adults. Although its precise etiology is unknown, low-grade inflammation in older adults is commonly associated with increased intestinal epithelial permeability (leaky gut) and abnormal (dysbiotic) gut microbiota. The increasing older population and lack of treatments to reduce aging-related microbiota dysbiosis, leaky gut, and inflammation culminates in a rise in aging-related comorbidities, constituting a significant public health concern. Here, we demonstrate that a human-origin probiotic cocktail containing 5 Lactobacillus and 5 Enterococcus strains isolated from healthy infant gut prevented high-fat diet-induced (HFD-induced) microbiota dysbiosis, leaky gut, inflammation, metabolic dysfunctions, and physical function decline in older mice. Probiotic-modulated gut microbiota primarily reduced leaky gut by increasing tight junctions, which in turn reduced inflammation. Mechanistically, probiotics modulated microbiota in a way to increase bile salt hydrolase activity, which in turn increased taurine abundance in the gut that stimulated tight junctions and suppressed gut leakiness. Furthermore, in Caenorhabditis elegans, taurine increased life span, reduced adiposity and leaky gut, and enhanced physical function. The results suggest that such probiotic therapies could prevent or treat aging-related leaky gut and inflammation in the elderly.
Collapse
Affiliation(s)
- Shokouh Ahmadi
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Shaohua Wang
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Ravinder Nagpal
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Bo Wang
- Department of Chemistry, North Carolina A&T State University, Greensboro, North Carolina, USA
| | - Shalini Jain
- Department of Internal Medicine-Endocrinology and Metabolism.,Mouse Metabolic Phenotyping Core
| | - Atefeh Razazan
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Sidharth P Mishra
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Xuewei Zhu
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA.,Department of Microbiology and Immunology, and
| | - Zhan Wang
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Kylie Kavanagh
- Department of Pathology-Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA.,Biomedical Sciences, University of Tasmania, Hobart, Australia
| | - Hariom Yadav
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA.,Department of Microbiology and Immunology, and
| |
Collapse
|