1
|
Patterson A, Boyle N, John J, Wang M, Mannochio-Russo H, Pyo JJ, Kim MS, Tian S, Koo I, Anitha M, Tian Y, Morgan E, Murray I, Perdew G, Wu G, Dorrestein P, Bisanz J, Redinbo M. Glucuronidation Metabolomic Fingerprinting to Map Host-Microbe Metabolism. RESEARCH SQUARE 2025:rs.3.rs-6321321. [PMID: 40297700 PMCID: PMC12036448 DOI: 10.21203/rs.3.rs-6321321/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Glucuronidation is an important detoxification pathway that operates in balance with gastrointestinal microbial β-glucuronidase (GUS) enzymes that can regenerate active metabolites from their glucuronidated forms. Although significant progress has been made in characterizing GUS enzymes, methods to comprehensively define the glucuronidome - the collection of glucuronidated metabolites - remain limited. In this study we employed pattern-filtering data science approaches alongside untargeted LC-MS/MS metabolomics to map the glucuronidome in urine, serum, and colon/fecal samples from gnotobiotic and conventional mice. Our findings reveal microbiome-driven shifts in the glucuronidome, highlighting how differential GUS activity can influence host metabolite profiles. Reverse metabolomics of known glucuronidated chemicals and glucuronidation pattern filtering searches in public metabolomics datasets exposed the diversity of glucuronidated metabolites in human and mouse ecosystems. In summary, we present a new glucuronidation fingerprint resource that provides broader access to and analysis of the glucuronidome. By systematically capturing glucuronidation patterns, this resource enhances unknown metabolite annotation efforts and provides new insights into the dynamic relationship between the host and bacterial biotransformation activities.
Collapse
|
2
|
Zhang R, Ding N, Feng X, Liao W. The gut microbiome, immune modulation, and cognitive decline: insights on the gut-brain axis. Front Immunol 2025; 16:1529958. [PMID: 39911400 PMCID: PMC11794507 DOI: 10.3389/fimmu.2025.1529958] [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: 11/20/2024] [Accepted: 01/07/2025] [Indexed: 02/07/2025] Open
Abstract
The gut microbiome has emerged as a pivotal area of research due to its significant influence on the immune system and cognitive functions. Cognitive disorders, including dementia and Parkinson's disease, represent substantial global health challenges. This review explores the relationship between gut microbiota, immune modulation, and cognitive decline, with a particular focus on the gut-brain axis. Research indicates that gut bacteria produce metabolites, including short-chain fatty acids (SCFAs), which affect mucosal immunity, antigen presentation, and immune responses, thereby influencing cognitive functions. A noteworthy correlation has been identified between imbalances in the gut microbiome and cognitive impairments, suggesting novel pathways for the treatment of cognitive disorders. Additionally, factors such as diet, environment, and pharmaceuticals play a role in shaping the composition of the gut microbiome, subsequently impacting both immune and cognitive health. This article aims to clarify the complex interactions among gut microbiota, immune regulation, and cognitive disorders, evaluating their potential as therapeutic targets. The goal is to promote microbiome-based treatments and lay the groundwork for future research in this field.
Collapse
Affiliation(s)
- Ruyi Zhang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
- Basic Medical School, Hubei University of Science and Technology, Xianning, China
| | - Ning Ding
- Basic Medical School, Hubei University of Science and Technology, Xianning, China
| | - Xicui Feng
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Wenli Liao
- Basic Medical School, Hubei University of Science and Technology, Xianning, China
| |
Collapse
|
3
|
Chen P. Targeting gut microbiota to counteract acetaminophen-induced acute liver injury. Trends Microbiol 2024; 32:419-421. [PMID: 38472076 DOI: 10.1016/j.tim.2024.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024]
Abstract
Acetaminophen (N-acetyl-p-aminophenol; APAP) overdose-induced acute liver injury (AILI) is a huge threat to public health worldwide. Recent research clearly shows that the intestinal microbiota (IM) is a key modulator in AILI. Herein, I discuss the latest findings on how the IM regulates AILI and the potential interventions to combat AILI by targeting the IM.
Collapse
Affiliation(s)
- Peng Chen
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
4
|
Chen X, Wu H, Li P, Peng W, Wang Y, Zhang X, Zhang A, Li J, Meng F, Wang W, Su W. Unraveling the Mechanism of Xiaochaihu Granules in Alleviating Yeast-Induced Fever Based on Network Analysis and Experimental Validation. Pharmaceuticals (Basel) 2024; 17:475. [PMID: 38675434 PMCID: PMC11053540 DOI: 10.3390/ph17040475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 03/30/2024] [Accepted: 04/06/2024] [Indexed: 04/28/2024] Open
Abstract
Xiaochaihu granules (XCHG) are extensively used to treat fever. Nevertheless, the underlying mechanism remains elusive. This study aimed to explore the potential of XCHG in mitigating yeast-induced fever and the underlying metabolic pathways. The chemical composition of XCHG was ascertained using ultra-fast liquid chromatography/quadrupole-time-of-flight tandem mass spectrometry (UFLC-Q-TOF-MS/MS), followed by integrated network analysis to predict potential targets. We then conducted experimental validation using pharmacological assays and metabolomics analysis in a yeast-induced mouse fever model. The study identified 133 compounds in XCHG, resulting in the development of a comprehensive network of herb-compound-biological functional modules. Subsequently, molecular dynamic (MD) simulations confirmed the stability of the complexes, including γ-aminobutyric acid B receptor 2 (GABBR2)-saikosaponin C, prostaglandin endoperoxide synthases (PTGS2)-lobetyolin, and NF-κB inhibitor IκBα (NFKBIA)-glycyrrhizic acid. Animal experiments demonstrated that XCHG reduced yeast-induced elevation in NFKBIA's downstream regulators [interleukin (IL)-1β and IL-8], inhibited PTGS2 activity, and consequently decreased prostaglandin E2 (PGE2) levels. XCHG also downregulated the levels of 5-hydroxytryptamine (5-HT), γ-aminobutyric acid (GABA), corticotropin releasing hormone (CRH), and adrenocorticotrophin (ACTH). These corroborated the network analysis results indicating XCHG's effectiveness against fever in targeting NFKBIA, PTGS2, and GABBR2. The hypothalamus metabolomics analysis identified 14 distinct metabolites as potential antipyretic biomarkers of XCHG. In conclusion, our findings suggest that XCHG alleviates yeast-induced fever by regulating inflammation/immune responses, neuromodulation, and metabolism modules, providing a scientific basis for the anti-inflammatory and antipyretic properties of XCHG.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Weiwei Su
- Guangdong Engineering & Technology Research Center for Quality and Efficacy Reevaluation of Post-Market Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Plant Resources, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| |
Collapse
|
5
|
Verdegaal AA, Goodman AL. Integrating the gut microbiome and pharmacology. Sci Transl Med 2024; 16:eadg8357. [PMID: 38295186 DOI: 10.1126/scitranslmed.adg8357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 01/11/2024] [Indexed: 02/02/2024]
Abstract
The gut microbiome harbors trillions of organisms that contribute to human health and disease. These bacteria can also affect the properties of medical drugs used to treat these diseases, and drugs, in turn, can reshape the microbiome. Research addressing interdependent microbiome-host-drug interactions thus has broad impact. In this Review, we discuss these interactions from the perspective of drug bioavailability, absorption, metabolism, excretion, toxicity, and drug-mediated microbiome modulation. We survey approaches that aim to uncover the mechanisms underlying these effects and opportunities to translate this knowledge into new strategies to improve the development, administration, and monitoring of medical drugs.
Collapse
Affiliation(s)
- Andrew A Verdegaal
- Department of Microbial Pathogenesis and Microbial Sciences Institute, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Andrew L Goodman
- Department of Microbial Pathogenesis and Microbial Sciences Institute, Yale University School of Medicine, New Haven, CT 06536, USA
| |
Collapse
|
6
|
Jiang DQY, Guo TL. Interaction between Per- and Polyfluorinated Substances (PFAS) and Acetaminophen in Disease Exacerbation-Focusing on Autism and the Gut-Liver-Brain Axis. TOXICS 2024; 12:39. [PMID: 38250995 PMCID: PMC10818890 DOI: 10.3390/toxics12010039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 12/05/2023] [Accepted: 12/22/2023] [Indexed: 01/23/2024]
Abstract
This review presents a new perspective on the exacerbation of autism spectrum disorder (ASD) by per- and polyfluoroalkyl substances (PFAS) through the gut-liver-brain axis. We have summarized evidence reported on the involvement of the gut microbiome and liver inflammation that led to the onset and exacerbation of ASD symptoms. As PFAS are toxicants that particularly target liver, this review has comprehensively explored the possible interaction between PFAS and acetaminophen, another liver toxicant, as the chemicals of interest for future toxicology research. Our hypothesis is that, at acute dosages, acetaminophen has the ability to aggravate the impaired conditions of the PFAS-exposed liver, which would further exacerbate neurological symptoms such as lack of social communication and interest, and repetitive behaviors using mechanisms related to the gut-liver-brain axis. This review discusses their potential interactions in terms of the gut-liver-brain axis and signaling pathways that may contribute to neurological diseases.
Collapse
Affiliation(s)
| | - Tai Liang Guo
- Department of Veterinary Biomedical Sciences, University of Georgia, Athens, GA 30602, USA;
| |
Collapse
|
7
|
Cai J, Auster A, Cho S, Lai Z. Dissecting the human gut microbiome to better decipher drug liability: A once-forgotten organ takes center stage. J Adv Res 2023; 52:171-201. [PMID: 37419381 PMCID: PMC10555929 DOI: 10.1016/j.jare.2023.07.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 05/25/2023] [Accepted: 07/03/2023] [Indexed: 07/09/2023] Open
Abstract
BACKGROUND The gut microbiome is a diverse system within the gastrointestinal tract composed of trillions of microorganisms (gut microbiota), along with their genomes. Accumulated evidence has revealed the significance of the gut microbiome in human health and disease. Due to its ability to alter drug/xenobiotic pharmacokinetics and therapeutic outcomes, this once-forgotten "metabolic organ" is receiving increasing attention. In parallel with the growing microbiome-driven studies, traditional analytical techniques and technologies have also evolved, allowing researchers to gain a deeper understanding of the functional and mechanistic effects of gut microbiome. AIM OF REVIEW From a drug development perspective, microbial drug metabolism is becoming increasingly critical as new modalities (e.g., degradation peptides) with potential microbial metabolism implications emerge. The pharmaceutical industry thus has a pressing need to stay up-to-date with, and continue pursuing, research efforts investigating clinical impact of the gut microbiome on drug actions whilst integrating advances in analytical technology and gut microbiome models. Our review aims to practically address this need by comprehensively introducing the latest innovations in microbial drug metabolism research- including strengths and limitations, to aid in mechanistically dissecting the impact of the gut microbiome on drug metabolism and therapeutic impact, and to develop informed strategies to address microbiome-related drug liability and minimize clinical risk. KEY SCIENTIFIC CONCEPTS OF REVIEW We present comprehensive mechanisms and co-contributing factors by which the gut microbiome influences drug therapeutic outcomes. We highlight in vitro, in vivo, and in silico models for elucidating the mechanistic role and clinical impact of the gut microbiome on drugs in combination with high-throughput, functionally oriented, and physiologically relevant techniques. Integrating pharmaceutical knowledge and insight, we provide practical suggestions to pharmaceutical scientists for when, why, how, and what is next in microbial studies for improved drug efficacy and safety, and ultimately, support precision medicine formulation for personalized and efficacious therapies.
Collapse
Affiliation(s)
- Jingwei Cai
- Drug Metabolism & Pharmacokinetics, Genentech Inc., South San Francisco, CA 94080, USA.
| | - Alexis Auster
- Drug Metabolism & Pharmacokinetics, Genentech Inc., South San Francisco, CA 94080, USA
| | - Sungjoon Cho
- Drug Metabolism & Pharmacokinetics, Genentech Inc., South San Francisco, CA 94080, USA
| | - Zijuan Lai
- Drug Metabolism & Pharmacokinetics, Genentech Inc., South San Francisco, CA 94080, USA
| |
Collapse
|
8
|
Nithiyanandam S, Evan Prince S. Caesalpinia bonducella mitigates oxidative damage by paracetamol intoxication in the kidney and intestine via modulating pro/anti-inflammatory and apoptotic signaling: an In vivo mechanistic insight. 3 Biotech 2023; 13:176. [PMID: 37188289 PMCID: PMC10175523 DOI: 10.1007/s13205-023-03601-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 04/29/2023] [Indexed: 05/17/2023] Open
Abstract
Protracted use of paracetamol at therapeutic/toxic doses readily induces major organ toxicity and poor clinical efficacy. Caesalpinia bonducella seeds possess a diverse range of biological and therapeutic activities. Thus, our study aimed to scrutinize the toxic effects of paracetamol and the potential renal and intestinal protective effects of Caesalpinia bonducella seed extract (CBSE). To Wistar rats, CBSE was administered for 8 days (300 mg/kg, p.o.) with or without paracetamol (2000 mg/kg, p.o.) on the 8th day. Pertinent toxicity assessments in the kidney and intestine were analyzed at the end of the study. The CBASE's phytochemical components were examined using gas chromatography-mass spectrometry (GC-MS). After the study period, study findings evidenced that paracetamol intoxication induced elevation of renal enzyme indicators, oxidative damage, imbalance with the pro/anti-inflammatory production and pro/anti-apoptotic mediators, and tissue injury; all repercussions were alleviated by pre-treatment with CBASE. CBASE considerably reduced (P < 0.05) paracetamol-induced kidney and intestine injury by limiting caspase-8/3 signaling and amplification of inflammation in renal and intestinal tissue by significantly reducing pro-inflammatory cytokine production. As per the GC-MS report, three main bioactive components-Piperine, Isocaryophyllene, and Tetradec-13-en-11-yn-1-ol were predominant and have protective activities. Our study ascertains that CBSE pre-treatment exerts potent renal and intestine protection against paracetamol intoxication. Thus, CBSE could be a prospective therapeutic candidate for protecting the kidney and intestine from the severity of paracetamol intoxication.
Collapse
Affiliation(s)
- Sangeetha Nithiyanandam
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, 632014 Tamil Nadu India
| | - Sabina Evan Prince
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, 632014 Tamil Nadu India
| |
Collapse
|
9
|
Džidić-Krivić A, Kusturica J, Sher EK, Selak N, Osmančević N, Karahmet Farhat E, Sher F. Effects of intestinal flora on pharmacokinetics and pharmacodynamics of drugs. Drug Metab Rev 2023; 55:126-139. [PMID: 36916327 DOI: 10.1080/03602532.2023.2186313] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Gut microbiota is known as unique collection of microorganisms (including bacteria, archaea, eukaryotes and viruses) that exist in a complex environment of the gut. Recently, this has become one of the most popular areas of research in medicine because this plays not only an important role in disease development, but gut microbiota also influences drug pharmacokinetics. These alterations in drug pharmacokinetic pathways and drug concentration in plasma and blood often lead to an increase in the incidence of toxicological events in patients. This review aims to present current knowledge of the most commonly used drugs in clinical practice and their dynamic interplay with the host's gut microbiota as well as the mechanisms underlying these metabolic processes and the consequent effect on their therapeutic efficacy and safety. These new findings set a foundation for the development of personalized treatments specific to each metabolism, maximizing drugs' therapeutic effects and minimizing the side effects because they are one of the major limiting factors in treating patients.
Collapse
Affiliation(s)
- Amina Džidić-Krivić
- Zenica Cantonal Hospital, Zenica, Bosnia and Herzegovina.,International Society of Engineering Science and Technology, Nottingham, UK
| | - Jasna Kusturica
- Faculty of Medicine, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Emina Karahmet Sher
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Nejra Selak
- Dom zdravlja Zenica, Zenica, Bosnia and Herzegovina
| | | | - Esma Karahmet Farhat
- International Society of Engineering Science and Technology, Nottingham, UK.,Department of Food and Nutrition Research, Faculty of Food Technology, Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Farooq Sher
- Department of Engineering, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| |
Collapse
|
10
|
Dichamp J, Cellière G, Ghallab A, Hassan R, Boissier N, Hofmann U, Reinders J, Sezgin S, Zühlke S, Hengstler JG, Drasdo D. In vitro to in vivo acetaminophen hepatotoxicity extrapolation using classical schemes, pharmacodynamic models and a multiscale spatial-temporal liver twin. Front Bioeng Biotechnol 2023; 11:1049564. [PMID: 36815881 PMCID: PMC9932319 DOI: 10.3389/fbioe.2023.1049564] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 01/10/2023] [Indexed: 02/05/2023] Open
Abstract
In vitro to in vivo extrapolation represents a critical challenge in toxicology. In this paper we explore extrapolation strategies for acetaminophen (APAP) based on mechanistic models, comparing classical (CL) homogeneous compartment pharmacodynamic (PD) models and a spatial-temporal (ST), multiscale digital twin model resolving liver microarchitecture at cellular resolution. The models integrate consensus detoxification reactions in each individual hepatocyte. We study the consequences of the two model types on the extrapolation and show in which cases these models perform better than the classical extrapolation strategy that is based either on the maximal drug concentration (Cmax) or the area under the pharmacokinetic curve (AUC) of the drug blood concentration. We find that an CL-model based on a well-mixed blood compartment is sufficient to correctly predict the in vivo toxicity from in vitro data. However, the ST-model that integrates more experimental information requires a change of at least one parameter to obtain the same prediction, indicating that spatial compartmentalization may indeed be an important factor.
Collapse
Affiliation(s)
- Jules Dichamp
- Group SIMBIOTX, INRIA Saclay-Île-de-France, Palaiseau, France,Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Dortmund, Germany,Group MAMBA, INRIA Paris, Paris, France
| | | | - Ahmed Ghallab
- Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Dortmund, Germany,Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Reham Hassan
- Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Dortmund, Germany,Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Noemie Boissier
- Group SIMBIOTX, INRIA Saclay-Île-de-France, Palaiseau, France
| | - Ute Hofmann
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology and University of Tübingen, Stuttgart, Germany
| | - Joerg Reinders
- Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Dortmund, Germany
| | - Selahaddin Sezgin
- Faculty of Chemistry and Chemical Biology, TU Dortmund, Dortmund, Germany
| | - Sebastian Zühlke
- Center for Mass Spectrometry (CMS), Faculty of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
| | - Jan G. Hengstler
- Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Dortmund, Germany
| | - Dirk Drasdo
- Group SIMBIOTX, INRIA Saclay-Île-de-France, Palaiseau, France,Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Dortmund, Germany,Group MAMBA, INRIA Paris, Paris, France,*Correspondence: Dirk Drasdo,
| |
Collapse
|
11
|
The past, present, and future of chemotherapy with a focus on individualization of drug dosing. J Control Release 2022; 352:840-860. [PMID: 36334860 DOI: 10.1016/j.jconrel.2022.10.043] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 10/14/2022] [Accepted: 10/24/2022] [Indexed: 11/16/2022]
Abstract
While there have been rapid advances in developing new and more targeted drugs to treat cancer, much less progress has been made in individualizing dosing. Even though the introduction of immunotherapies such as CAR T-cells and checkpoint inhibitors, as well as personalized therapies that target specific mutations, have transformed clinical treatment of cancers, chemotherapy remains a mainstay in oncology. Chemotherapies are typically dosed on either a body surface area (BSA) or weight basis, which fails to account for pharmacokinetic differences between patients. Drug absorption, distribution, metabolism, and excretion rates can vary between patients, resulting in considerable differences in exposure to the active drugs. These differences result in suboptimal dosing, which can reduce efficacy and increase side-effects. Therapeutic drug monitoring (TDM), genotype guided dosing, and chronomodulation have been developed to address this challenge; however, despite improving clinical outcomes, they are rarely implemented in clinical practice for chemotherapies. Thus, there is a need to develop interventions that allow for individualized drug dosing of chemotherapies, which can help maximize the number of patients that reach the most efficacious level of drug in the blood while mitigating the risks of underdosing or overdosing. In this review, we discuss the history of the development of chemotherapies, their mechanisms of action and how they are dosed. We discuss substantial intraindividual and interindividual variability in chemotherapy pharmacokinetics. We then propose potential engineering solutions that could enable individualized dosing of chemotherapies, such as closed-loop drug delivery systems and bioresponsive biomaterials.
Collapse
|
12
|
Laue HE, Shen Y, Bloomquist TR, Wu H, Brennan KJM, Cassoulet R, Wilkie E, Gillet V, Desautels AS, Abdelouahab N, Bellenger JP, Burris HH, Coull BA, Weisskopf MG, Zhang W, Takser L, Baccarelli AA. In Utero Exposure to Caffeine and Acetaminophen, the Gut Microbiome, and Neurodevelopmental Outcomes: A Prospective Birth Cohort Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:9357. [PMID: 35954712 PMCID: PMC9367926 DOI: 10.3390/ijerph19159357] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/22/2022] [Accepted: 07/28/2022] [Indexed: 02/05/2023]
Abstract
Pregnant individuals are exposed to acetaminophen and caffeine, but it is unknown how these exposures interact with the developing gut microbiome. We aimed to determine whether acetaminophen and/or caffeine relate to the childhood gut microbiome and whether features of the gut microbiome alter the relationship between acetaminophen/caffeine and neurodevelopment. Forty-nine and 85 participants provided meconium and stool samples at 6-7, respectively, for exposure and microbiome assessment. Fecal acetaminophen and caffeine concentrations were quantified, and fecal DNA underwent metagenomic sequencing. Caregivers and study staff assessed the participants' motor and cognitive development using standardized scales. Prenatal exposures had stronger associations with the childhood microbiome than concurrent exposures. Prenatal acetaminophen exposure was associated with a trend of lower gut bacterial diversity in childhood [β = -0.17 Shannon Index, 95% CI: (-0.31, -0.04)] and was marginally associated with differences in the relative abundances of features of the gut microbiome at the phylum (Firmicutes, Actinobacteria) and gene pathway levels. Among the participants with a higher relative abundance of Proteobacteria, prenatal exposure to acetaminophen and caffeine was associated with lower scores on WISC-IV subscales. Acetaminophen during bacterial colonization of the naïve gut is associated with lasting alterations in childhood microbiome composition. Future studies may inform our understanding of downstream health effects.
Collapse
Affiliation(s)
- Hannah E. Laue
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA
| | - Yike Shen
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY 10032, USA; (Y.S.); (T.R.B.); (H.W.); (K.J.M.B.); (A.A.B.)
| | - Tessa R. Bloomquist
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY 10032, USA; (Y.S.); (T.R.B.); (H.W.); (K.J.M.B.); (A.A.B.)
| | - Haotian Wu
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY 10032, USA; (Y.S.); (T.R.B.); (H.W.); (K.J.M.B.); (A.A.B.)
| | - Kasey J. M. Brennan
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY 10032, USA; (Y.S.); (T.R.B.); (H.W.); (K.J.M.B.); (A.A.B.)
| | - Raphael Cassoulet
- Département de Chimie, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (R.C.); (J.P.B.)
| | - Erin Wilkie
- Département de Pédiatrie, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (E.W.); (V.G.); (A.-S.D.); (N.A.); (L.T.)
| | - Virginie Gillet
- Département de Pédiatrie, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (E.W.); (V.G.); (A.-S.D.); (N.A.); (L.T.)
| | - Anne-Sandrine Desautels
- Département de Pédiatrie, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (E.W.); (V.G.); (A.-S.D.); (N.A.); (L.T.)
| | - Nadia Abdelouahab
- Département de Pédiatrie, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (E.W.); (V.G.); (A.-S.D.); (N.A.); (L.T.)
| | - Jean Philippe Bellenger
- Département de Chimie, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (R.C.); (J.P.B.)
| | - Heather H. Burris
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
- Division of Neonatology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Brent A. Coull
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA;
| | - Marc G. Weisskopf
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA;
| | - Wei Zhang
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA;
| | - Larissa Takser
- Département de Pédiatrie, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (E.W.); (V.G.); (A.-S.D.); (N.A.); (L.T.)
- Département de Psychiatrie, Faculté de Médicine et Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada
| | - Andrea A. Baccarelli
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY 10032, USA; (Y.S.); (T.R.B.); (H.W.); (K.J.M.B.); (A.A.B.)
| |
Collapse
|
13
|
Yu F, Liu Y, Zhang R, Zhu L, Zhang T, Shi Y. Recent advances in circadian-regulated pharmacokinetics and its implications for chronotherapy. Biochem Pharmacol 2022; 203:115185. [PMID: 35902039 DOI: 10.1016/j.bcp.2022.115185] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/19/2022] [Accepted: 07/19/2022] [Indexed: 11/02/2022]
Abstract
Dependence of pharmacokinetics and drug effects (efficacy and toxicity) on dosing time has long been recognized. However, significant progress has only recently been made in our understanding of circadian rhythms and their regulation on drug pharmacokinetics, efficacy and toxicity. This review will cover the relevant literature and a series of publications from our work summarizing the effects of circadian rhythms on drug pharmacokinetics, and propose that the influence of circadian rhythms on pharmacokinetics are ultimately translated into therapeutic effects and side effects of drugs. Evidence suggests that daily rhythmicity in expression of drug-metabolizing enzymes and transporters necessary for drug ADME (absorption, distribution, metabolism and excretion) are key factors determining circadian pharmacokinetics. Newly discovered mechanisms for circadian control of the enzymes and transporters are covered. We also discuss how the rhythms of drug-processing proteins are translated into circadian pharmacokinetics and drug chronoefficacy/chronotoxicity, which has direct implications for chronotherapy. More importantly, we will present perspectives on the challenges that are still needed for a breakthrough in translational research. In addition, knowledge of the circadian influence on drug disposition has provided new possibilities for novel pharmacological strategies. Careful application of pharmacokinetics-based chronotherapy strategies can improve efficacy and reduce toxicity. Circadian rhythm-mediated metabolic and transport strategies can also be implemented to design drugs.
Collapse
Affiliation(s)
- Fangjun Yu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuanyuan Liu
- School of Fundamental Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Rong Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lijun Zhu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tianpeng Zhang
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Yafei Shi
- School of Fundamental Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
14
|
Tesei D, Jewczynko A, Lynch AM, Urbaniak C. Understanding the Complexities and Changes of the Astronaut Microbiome for Successful Long-Duration Space Missions. Life (Basel) 2022; 12:life12040495. [PMID: 35454986 PMCID: PMC9031868 DOI: 10.3390/life12040495] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/17/2022] [Accepted: 03/24/2022] [Indexed: 12/12/2022] Open
Abstract
During space missions, astronauts are faced with a variety of challenges that are unique to spaceflight and that have been known to cause physiological changes in humans over a period of time. Several of these changes occur at the microbiome level, a complex ensemble of microbial communities residing in various anatomic sites of the human body, with a pivotal role in regulating the health and behavior of the host. The microbiome is essential for day-to-day physiological activities, and alterations in microbiome composition and function have been linked to various human diseases. For these reasons, understanding the impact of spaceflight and space conditions on the microbiome of astronauts is important to assess significant health risks that can emerge during long-term missions and to develop countermeasures. Here, we review various conditions that are caused by long-term space exploration and discuss the role of the microbiome in promoting or ameliorating these conditions, as well as space-related factors that impact microbiome composition. The topics explored pertain to microgravity, radiation, immunity, bone health, cognitive function, gender differences and pharmacomicrobiomics. Connections are made between the trifecta of spaceflight, the host and the microbiome, and the significance of these interactions for successful long-term space missions.
Collapse
Affiliation(s)
- Donatella Tesei
- Department of Biotechnology, University of Natural Resources and Life Sciences, 1190 Vienna, Austria;
| | - Anna Jewczynko
- Department of Biology, University of Waterloo, Waterloo, ON N2L 3G1, Canada;
| | - Anne M. Lynch
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Camilla Urbaniak
- ZIN Technologies Inc., Middleburg Heights, OH 44130, USA
- NASA Jet Propulsion Laboratory, California Institute of Technology, Pasadena, CA 91109, USA
- Correspondence:
| |
Collapse
|
15
|
Bai X, Liu G, Yang J, Zhu J, Li X. Gut Microbiota as the Potential Mechanism to Mediate Drug Metabolism Under High-Altitude Hypoxia. Curr Drug Metab 2022; 23:8-20. [PMID: 35088664 DOI: 10.2174/1389200223666220128141038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/25/2021] [Accepted: 12/30/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND The characteristics of pharmacokinetics and the activity and expression of drug-metabolizing enzymes and transporters significantly change under a high-altitude hypoxic environment. Gut microbiota is an important factor affecting the metabolism of drugs through direct or indirect effects, changing the bioavailability, biological activity, or toxicity of drugs and further affecting the efficacy and safety of drugs in vivo. A high-altitude hypoxic environment significantly changes the structure and diversity of gut microbiota, which may play a key role in drug metabolism under a high-altitude hypoxic environment. METHODS An investigation was carried out by reviewing published studies to determine the role of gut microbiota in the regulation of drug-metabolizing enzymes and transporters. Data and information on expression change in gut microbiota, drug-metabolizing enzymes and transporters under a high-altitude hypoxic environment were explored and proposed. RESULTS High-altitude hypoxia is an important environmental factor that can adjust the structure of the gut microbiota and change the diversity of intestinal microbes. It was speculated that the gut microbiota could regulate drug-metabolizing enzymes through two potential mechanisms, the first being through direct regulation of the metabolism of drugs in vivo and the second being indirect, i.e., through the regulation of drug-metabolizing enzymes and transporters, thereby affecting the activity of drugs. CONCLUSION This article reviews the effects of high-altitude hypoxia on the gut microbiota and the effects of these changes on drug metabolism.
Collapse
Affiliation(s)
- Xue Bai
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining, China
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, China
| | - Guiqin Liu
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining, China
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, China
| | - Jianxin Yang
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining, China
| | - Junbo Zhu
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining, China
| | - Xiangyang Li
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, China
| |
Collapse
|
16
|
Tsunoda SM, Gonzales C, Jarmusch AK, Momper JD, Ma JD. Contribution of the Gut Microbiome to Drug Disposition, Pharmacokinetic and Pharmacodynamic Variability. Clin Pharmacokinet 2021; 60:971-984. [PMID: 33959897 PMCID: PMC8332605 DOI: 10.1007/s40262-021-01032-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2021] [Indexed: 12/20/2022]
Abstract
The trillions of microbes that make up the gut microbiome are an important contributor to health and disease. With respect to xenobiotics, particularly orally administered compounds, the gut microbiome interacts directly with drugs to break them down into metabolic products. In addition, microbial products such as bile acids interact with nuclear receptors on host drug-metabolizing enzyme machinery, thus indirectly influencing drug disposition and pharmacokinetics. Gut microbes also influence drugs that undergo enterohepatic recycling by reversing host enzyme metabolic processes and increasing exposure to toxic metabolites as exemplified by the chemotherapy agent irinotecan and non-steroidal anti-inflammatory drugs. Recent data with immune checkpoint inhibitors demonstrate the impact of the gut microbiome on drug pharmacodynamics. We summarize the clinical importance of gut microbe interaction with digoxin, irinotecan, immune checkpoint inhibitors, levodopa, and non-steroidal anti-inflammatory drugs. Understanding the complex interactions of the gut microbiome with xenobiotics is challenging; and highly sensitive methods such as untargeted metabolomics with molecular networking along with other in silico methods and animal and human in vivo studies will uncover mechanisms and pathways. Incorporating the contribution of the gut microbiome to drug disposition, pharmacokinetics, and pharmacodynamics is vital in this era of precision medicine.
Collapse
Affiliation(s)
- Shirley M Tsunoda
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, MC 0657, La Jolla, San Diego, CA, 90293-0657, USA.
| | - Christopher Gonzales
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, MC 0657, La Jolla, San Diego, CA, 90293-0657, USA
| | - Alan K Jarmusch
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, MC 0657, La Jolla, San Diego, CA, 90293-0657, USA.,Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, San Diego, CA, USA
| | - Jeremiah D Momper
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, MC 0657, La Jolla, San Diego, CA, 90293-0657, USA
| | - Joseph D Ma
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, MC 0657, La Jolla, San Diego, CA, 90293-0657, USA
| |
Collapse
|
17
|
Hirosawa K, Fukami T, Nagaoka M, Nakano M, Nakajima M. Methionine Sulfoxide Reductase A in Human and Mouse Tissues is Responsible for Sulindac Activation, Making a Larger Contribution than the Gut Microbiota. Drug Metab Dispos 2021; 50:725-733. [PMID: 35279645 DOI: 10.1124/dmd.122.000828] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/01/2022] [Indexed: 02/13/2025] Open
Abstract
Sulindac is a nonsteroidal anti-inflammatory prodrug that is converted to its pharmacologically active metabolite, sulindac sulfide, via a reduction reaction. It is widely accepted that the gut microbiota is responsible for sulindac activation; however, sulindac-induced gastrointestinal injury, which is caused by irritation of the gastrointestinal tract by its active metabolite, is uncommon. Therefore, it is surmised that sulindac is converted to its active metabolite in tissues after absorption. In this study, we sought to identify the enzyme(s) responsible for sulindac activation in tissues and to compare its/their contribution to the gut microbiota. Sulindac is enzymatically reduced in human intestinal, liver, and renal cytosols. Since sulindac is known to be reduced by methionine sulfoxide reductase (Msr) in Escherichia coli, we investigated whether the human ortholog MSRA catalyzes the sulindac reduction reaction. We found that recombinant human MSRA shows sulindac reductase activity with a similar Michaelis constant value as tissue cytosols. In addition, it was revealed that cytosolic factor(s) efficiently enhanced MSRA activity. By using the relative expression factor, the contribution of MSRA to the sulindac reductase activities in each tissue cytosol was calculated to be almost 100%. In mice, depletion of the gut microbiota by administration of antibiotics resulted in a 31% decrease in the area under the curve ratio of sulindac sulfide to sulindac, indicating that the contribution of tissue MsrA to sulindac activation is expected to be 69% in the body. In conclusion, we demonstrated that MSRA expressed in tissues is involved in sulindac activation, making a larger contribution than the gut microbiota. SIGNIFICANCE STATEMENT: Methionine sulfoxide reductase A is responsible for the activation of sulindac, a nonsteroidal anti-inflammatory prodrug, to sulindac sulfide, an active form, in human tissues. Methionine sulfoxide reductase A expressed in tissues activates sulindac with a higher contribution than gut microbiota in body.
Collapse
Affiliation(s)
- Keiya Hirosawa
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan (K.H., M.Nag., Ma.Nak., Mi.Nak.) and WPI Nano Life Science Institute, Kanazawa, Japan (T.F., M.Nag., Mi.Nak.)
| | - Tatsuki Fukami
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan (K.H., M.Nag., Ma.Nak., Mi.Nak.) and WPI Nano Life Science Institute, Kanazawa, Japan (T.F., M.Nag., Mi.Nak.)
| | - Mai Nagaoka
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan (K.H., M.Nag., Ma.Nak., Mi.Nak.) and WPI Nano Life Science Institute, Kanazawa, Japan (T.F., M.Nag., Mi.Nak.)
| | - Masataka Nakano
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan (K.H., M.Nag., Ma.Nak., Mi.Nak.) and WPI Nano Life Science Institute, Kanazawa, Japan (T.F., M.Nag., Mi.Nak.)
| | - Miki Nakajima
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan (K.H., M.Nag., Ma.Nak., Mi.Nak.) and WPI Nano Life Science Institute, Kanazawa, Japan (T.F., M.Nag., Mi.Nak.)
| |
Collapse
|
18
|
Papageorgiou M, Biver E. Interactions of the microbiome with pharmacological and non-pharmacological approaches for the management of ageing-related musculoskeletal diseases. Ther Adv Musculoskelet Dis 2021; 13:1759720X211009018. [PMID: 34104230 PMCID: PMC8172340 DOI: 10.1177/1759720x211009018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/18/2021] [Indexed: 12/18/2022] Open
Abstract
Despite major progress in the understanding of the pathophysiology and therapeutic options for common ageing-related musculoskeletal conditions (i.e. osteoporosis and associated fractures, sarcopenia and osteoarthritis), there is still a considerable proportion of patients who respond sub optimally to available treatments or experience adverse effects. Emerging microbiome research suggests that perturbations in microbial composition, functional and metabolic capacity (i.e. dysbiosis) are associated with intestinal and extra-intestinal disorders including musculoskeletal diseases. Besides its contributions to disease pathogenesis, the role of the microbiome is further extended to shaping individuals' responses to disease therapeutics (i.e. pharmacomicrobiomics). In this review, we focus on the reciprocal interactions between the microbiome and therapeutics for osteoporosis, sarcopenia and osteoarthritis. Specifically, we identify the effects of therapeutics on microbiome's configurations, functions and metabolic output, intestinal integrity and immune function, but also the effects of the microbiome on the metabolism of these therapeutics, which in turn, may influence their bioavailability, efficacy and side-effect profile contributing to variable treatment responses in clinical practice. We further discuss emerging strategies for microbiota manipulation as preventive or therapeutic (alone or complementary to available treatments) approaches for improving outcomes of musculoskeletal health and disease.
Collapse
Affiliation(s)
- Maria Papageorgiou
- Division of Bone Diseases, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Emmanuel Biver
- Division of Bone Diseases, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Rue Gabrielle Perret Gentil 4, Geneva 1205, Switzerland
| |
Collapse
|
19
|
Circadian rhythms: influence on physiology, pharmacology, and therapeutic interventions. J Pharmacokinet Pharmacodyn 2021; 48:321-338. [PMID: 33797011 PMCID: PMC8015932 DOI: 10.1007/s10928-021-09751-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 03/19/2021] [Indexed: 12/20/2022]
Abstract
Circadian rhythms are ubiquitous phenomena that recur daily in a self-sustaining, entrainable, and oscillatory manner, and orchestrate a wide range of molecular, physiological, and behavioral processes. Circadian clocks are comprised of a hierarchical network of central and peripheral clocks that generate, sustain, and synchronize the circadian rhythms. The functioning of the peripheral clock is regulated by signals from autonomic innervation (from the central clock), endocrine networks, feeding, and other external cues. The critical role played by circadian rhythms in maintaining both systemic and tissue-level homeostasis is well established, and disruption of the rhythm has direct consequence for human health, disorders, and diseases. Circadian oscillations in both pharmacokinetics and pharmacodynamic processes are known to affect efficacy and toxicity of several therapeutic agents. A variety of modeling approaches ranging from empirical to more complex systems modeling approaches have been applied to characterize circadian biology and its influence on drug actions, optimize time of dosing, and identify opportunities for pharmacological modulation of the clock mechanisms and their downstream effects. In this review, we summarize current understanding of circadian rhythms and its influence on physiology, pharmacology, and therapeutic interventions, and discuss the role of chronopharmacometrics in gaining new insights into circadian rhythms and its applications in chronopharmacology.
Collapse
|
20
|
Zimmermann M, Patil KR, Typas A, Maier L. Towards a mechanistic understanding of reciprocal drug-microbiome interactions. Mol Syst Biol 2021; 17:e10116. [PMID: 33734582 PMCID: PMC7970330 DOI: 10.15252/msb.202010116] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/10/2021] [Accepted: 01/25/2021] [Indexed: 02/06/2023] Open
Abstract
Broad-spectrum antibiotics target multiple gram-positive and gram-negative bacteria, and can collaterally damage the gut microbiota. Yet, our knowledge of the extent of damage, the antibiotic activity spectra, and the resistance mechanisms of gut microbes is sparse. This limits our ability to mitigate microbiome-facilitated spread of antibiotic resistance. In addition to antibiotics, non-antibiotic drugs affect the human microbiome, as shown by metagenomics as well as in vitro studies. Microbiome-drug interactions are bidirectional, as microbes can also modulate drugs. Chemical modifications of antibiotics mostly function as antimicrobial resistance mechanisms, while metabolism of non-antibiotics can also change the drugs' pharmacodynamic, pharmacokinetic, and toxic properties. Recent studies have started to unravel the extensive capacity of gut microbes to metabolize drugs, the mechanisms, and the relevance of such events for drug treatment. These findings raise the question whether and to which degree these reciprocal drug-microbiome interactions will differ across individuals, and how to take them into account in drug discovery and precision medicine. This review describes recent developments in the field and discusses future study areas that will benefit from systems biology approaches to better understand the mechanistic role of the human gut microbiota in drug actions.
Collapse
Affiliation(s)
- Michael Zimmermann
- Structural and Computational Biology UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
| | - Kiran Raosaheb Patil
- Structural and Computational Biology UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
- The Medical Research Council Toxicology UnitUniversity of CambridgeCambridgeUK
| | - Athanasios Typas
- Structural and Computational Biology UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
- Genome Biology UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
| | - Lisa Maier
- Interfaculty Institute of Microbiology and Infection MedicineUniversity of TübingenTübingenGermany
- Cluster of Excellence ‘Controlling Microbes to Fight Infections’University of TübingenTübingenGermany
| |
Collapse
|