1
|
Langeraert J, Gasthuys E, Vermeulen A. Small molecule drug absorption in inflammatory bowel disease and current implementation in physiologically- based pharmacokinetic models. Eur J Pharm Sci 2025; 209:107095. [PMID: 40187540 DOI: 10.1016/j.ejps.2025.107095] [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: 08/12/2024] [Revised: 02/09/2025] [Accepted: 04/03/2025] [Indexed: 04/07/2025]
Abstract
Inflammatory bowel disease (IBD) is characterized by a chronic inflammation of the intestinal mucosa, with predominant localization in the colon in ulcerative colitis (UC) or affecting the entire length of the gastrointestinal tract in Crohn's disease (CD). Recent advances in the drug development space have been marked by a return to orally administered small molecules with novel mechanisms of action such as Janus kinase inhibitors. Additionally, the prevalence of certain chronic conditions is higher in IBD patients, many of which are treated with orally administered drugs. Given the pathophysiology and localization of IBD, altered drug absorption from the gastrointestinal tract can be expected. This review discusses several physiological differences between the small and large intestine with the potential to influence drug absorption including pathophysiology related alterations associated with IBD. The main physiological parameters which are identified include luminal fluid volume, luminal pH, transit time, bile salt concentration, microbiome, absorptive surface area, permeability and metabolizing enzymes and transporters. Literature regarding these factors in IBD patients is marked with high heterogeneity in reporting of disease severity and location leading to difficulties in interpreting data across different studies. While the influence of most of these factors has been directly assessed in healthy volunteers, this is rarely the case for IBD patients. Furthermore, studies which used PBPK modelling to describe the PK of an orally administered drug in an IBD population and were able to verify their findings using clinical data are critically examined. These models were able to incorporate the pathophysiological changes associated with IBD and partly succeeded in adequately predicting drug absorption in this population. Given the limited amount of PBPK studies performed on a limited number of drugs, the developed models are most likely not suitable to be used as a general PBPK model for the IBD population.
Collapse
Affiliation(s)
- Jonas Langeraert
- Laboratory of Medicinal Biochemistry and Clinical Analysis, Department of Bioanalysis, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| | - Elke Gasthuys
- Laboratory of Medicinal Biochemistry and Clinical Analysis, Department of Bioanalysis, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - An Vermeulen
- Laboratory of Medicinal Biochemistry and Clinical Analysis, Department of Bioanalysis, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| |
Collapse
|
2
|
Zhou J, Li TL, Wei B, Ruan YF, Wang YQ, Liu JY, Song MM, Shen YX. Oral colon-targeted delivery of recombinant human MANF for alleviation of ulcerative colitis. Int J Pharm X 2025; 9:100320. [PMID: 40115964 PMCID: PMC11925120 DOI: 10.1016/j.ijpx.2025.100320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 02/14/2025] [Accepted: 02/20/2025] [Indexed: 03/23/2025] Open
Abstract
Midbrain astrocyte-derived neurotrophic factor (MANF) is a secreted protein induced by endoplasmic reticulum stress. Previous studies have indicated that intravenous administration of 1 mg/kg/day recombinant human MANF protein with His tag (His-MANF) for 3 days can ameliorate acute ulcerative colitis in mice. However, long-term intravenous therapy has many disadvantages. In this paper, His-MANF protein was successfully encapsulated into alginate and hyaluronic acid hybrid hydrogel microcapsules in one step using the gas shear method and then coated by Eudragit S100 to construct an oral colon-targeted delivery system (MSH@E). The MSH@E microcapsules exhibited controlled and sustained release behavior and colon-targeting properties. Both fluorescent imaging and immunohistochemistry staining results showed that His-MANF protein could accumulate in the colitis colon for a longer residence time after oral delivery. In vivo studies demonstrated that oral administration of MSH@E microcapsules could alleviate DSS-induced colitis in mice without systemic toxicity. Importantly, even if the oral His-MANF dose was half of the intravenous His-MANF dose, oral delivery was still much more effective than intravenous injection, suggesting the development of the oral colon-targeted delivery system (MSH@E) has great significance and makes a breakthrough from intravenous to oral administration for His-MANF treatment of ulcerative colitis (UC).
Collapse
Affiliation(s)
- Jie Zhou
- School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, 230032 Hefei, Anhui, PR China
- Anhui Provincial Institute of Translational Medicine, 230032 Hefei, Anhui, PR China
| | - Tian-Le Li
- School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, 230032 Hefei, Anhui, PR China
| | - Bo Wei
- School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, 230032 Hefei, Anhui, PR China
- Anhui Provincial Institute of Translational Medicine, 230032 Hefei, Anhui, PR China
| | - Yue-Feng Ruan
- School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, 230032 Hefei, Anhui, PR China
- Anhui Provincial Institute of Translational Medicine, 230032 Hefei, Anhui, PR China
| | - Ye-Qin Wang
- School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, 230032 Hefei, Anhui, PR China
- Anhui Provincial Institute of Translational Medicine, 230032 Hefei, Anhui, PR China
| | - Jiao-Yan Liu
- School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, 230032 Hefei, Anhui, PR China
| | - Meng-Meng Song
- School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, 230032 Hefei, Anhui, PR China
| | - Yu-Xian Shen
- School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, 230032 Hefei, Anhui, PR China
- Anhui Provincial Institute of Translational Medicine, 230032 Hefei, Anhui, PR China
| |
Collapse
|
3
|
Petersen I, Jonusaite S, Thoben F, Hu MY. Evidence for HCO 3- and NH 3/NH 4+-dependent pH regulatory mechanisms in the alkaline midgut of the sea urchin larva. Am J Physiol Regul Integr Comp Physiol 2025; 328:R685-R699. [PMID: 40248920 DOI: 10.1152/ajpregu.00222.2024] [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: 09/10/2024] [Revised: 11/07/2024] [Accepted: 03/20/2025] [Indexed: 04/19/2025]
Abstract
Alkaline digestive systems are well described for some insect species and their larval stages. More recently, larvae of the members of ambulacraria superphylum consisting of echinoderms and hemichordates were also discovered to have highly alkaline midguts (pH 9.5-10.5) with the underlying acid-base regulatory mechanisms largely unknown. Using pharmacological inhibition of acid-base transporters in conjunction with ion-selective microelectrode measurements and pH-sensitive dyes, we investigated intracellular and extracellular pH regulatory mechanisms of midgut epithelial cells of a sea urchin (Strongylocentrotus purpuratus) larva. Our findings suggest that vacuolar-type H+-ATPase (inhibited by bafilomycin a1), carbonic anhydrase (inhibited by acetazolamide), anion-exchangers (inhibited by 4,4'-diisothiocyano-2,2'-disulfonic acid or DIDS), and soluble adenylyl cyclase (inhibited by KH7) play crucial roles in cellular acid-base regulation as well as midgut alkalization. Ammonia excretion rates were decreased in the presence of bafilomycin and colchicine, pointing toward vesicular [Formula: see text] trapping and exocytosis mechanism in eliminating nitrogenous proton equivalents from midgut cells. Finally, midgut perfusion studies revealed ouabain-sensitive luminal [Formula: see text] uptake, suggesting a role for Na+/K+-ATPase-mediated ammonia transport in midgut alkalization. This comprehensive pharmacological analysis provides a new working model relying on the CO2/[Formula: see text] and NH3/[Formula: see text] buffer systems for midgut alkalization in the sea urchin larva. These findings are discussed in the context of other alkalizing systems with strong implications for the conserved role of [Formula: see text] and NH3-driven mechanism of midgut alkalization across the animal kingdom.NEW & NOTEWORTHY Sea urchin larvae evolved highly alkaline conditions in their digestive tracts, and the underlying acid-base regulatory mechanisms are little understood. Here we present evidence that the process of luminal alkalization is cAMP-dependent. Furthermore, our data point toward the involvement of bicarbonate and ammonia in regulating midgut fluid pH. These results identified a novel mechanism for luminal alkalization in the digestive tract of a marine animal with strong implications for other alkalizing systems in animals.
Collapse
Affiliation(s)
- Inga Petersen
- Institute of Physiology, Christian-Albrechts University of Kiel, Kiel, Germany
| | - Sima Jonusaite
- Institute of Physiology, Christian-Albrechts University of Kiel, Kiel, Germany
- Department of Biological Sciences, University of Tulsa, Tulsa, Oklahoma, United States
| | - Femke Thoben
- Institute of Physiology, Christian-Albrechts University of Kiel, Kiel, Germany
| | - Marian Y Hu
- Institute of Physiology, Christian-Albrechts University of Kiel, Kiel, Germany
| |
Collapse
|
4
|
Adelfio M, Callen GE, He X, Paster BJ, Hasturk H, Ghezzi CE. Engineered Tissue Models to Decode Host-Microbiota Interactions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2417687. [PMID: 40364768 DOI: 10.1002/advs.202417687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 04/13/2025] [Indexed: 05/15/2025]
Abstract
A mutualistic co-evolution exists between the host and its associated microbiota in the human body. Bacteria establish ecological niches in various tissues of the body, locally influencing their physiology and functions, but also contributing to the well-being of the whole organism through systemic communication with other distant niches (axis). Emerging evidence indicates that when the composition of the microbiota inhabiting the niche changes toward a pathogenic state (dysbiosis) and interactions with the host become unbalanced, diseases may present. In addition, imbalances within a single niche can cause dysbiosis in distant organs. Current research efforts are focused on elucidating the mechanisms leading to dysbiosis, with the goal of restoring tissue homeostasis. In vitro models can provide critical experimental platforms to address this need, by reproducing the niche cyto-architecture and physiology with high fidelity. This review surveys current in in vitro host-microbiota research strategies and provides a roadmap that can guide the field in further developing physiologically relevant in vitro models of ecological niches, thus enabling investigation of the role of the microbiota in human health and diseases. Lastly, given the Food and Drug Administration Modernization Act 2.0, this review highlights emerging in vitro strategies to support the development and validation of new therapies on the market.
Collapse
Affiliation(s)
- Miryam Adelfio
- Department of Biomedical Engineering, University of Massachusetts-Lowell, Lowell, MA, 01854, USA
| | - Grace E Callen
- Department of Biomedical Engineering, University of Massachusetts-Lowell, Lowell, MA, 01854, USA
| | - Xuesong He
- ADA Forsyth Institute, 245 First St, Cambridge, MA, 02142, USA
| | - Bruce J Paster
- ADA Forsyth Institute, 245 First St, Cambridge, MA, 02142, USA
| | - Hatice Hasturk
- ADA Forsyth Institute, 245 First St, Cambridge, MA, 02142, USA
| | - Chiara E Ghezzi
- Department of Biomedical Engineering, University of Massachusetts-Lowell, Lowell, MA, 01854, USA
| |
Collapse
|
5
|
Gelli HP, Vazquez-Uribe R, Buckley ST, Andersen JT, Alexander Sommer MO. Advanced microbiome therapeutics for oral delivery of peptides and proteins: Advances, challenges, and opportunities. Adv Drug Deliv Rev 2025; 222:115603. [PMID: 40349728 DOI: 10.1016/j.addr.2025.115603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2025] [Revised: 05/01/2025] [Accepted: 05/06/2025] [Indexed: 05/14/2025]
Abstract
Peptide and protein medicines have changed the therapeutic landscape for many diseases, yet oral delivery remains a significant challenge due to enzymatic degradation, instability, and poor permeability in the gastrointestinal tract. Advanced Microbiome Therapeutics (AMTs) could overcome some of these barriers by producing and releasing therapeutic peptides directly in the gastrointestinal tract. AMTs can localize peptide production at the site of absorption, providing either sustained or controlled release while potentially reducing side effects associated with systemic administration. Here, this review assesses the status of AMTs for oral peptide delivery and discusses the potential integration of permeation enhancers, mucoadhesive systems, and receptor-mediated transport strategies to improve oral bioavailability further. Combining these approaches could pave the way for more widespread oral delivery strategies for peptide and protein medicines.
Collapse
Affiliation(s)
- Hitesh P Gelli
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | | | | | - Jan Terje Andersen
- Department of Pharmacology, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo, Norway; Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
| | | |
Collapse
|
6
|
Vinarov Z, Müllertz A, Mircheva H, Gouar YL, Menard O, Kumar SP, Paudel A, Dupont D, Augustijns P. Intraluminal enzymatic hydrolysis of API and lipid or polymeric excipients. Int J Pharm 2025; 675:125489. [PMID: 40154815 DOI: 10.1016/j.ijpharm.2025.125489] [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: 01/23/2025] [Revised: 03/14/2025] [Accepted: 03/15/2025] [Indexed: 04/01/2025]
Abstract
The role of intraluminal enzymes for the hydrolysis of active pharmaceutical ingredients (API), prodrugs and pharmaceutical excipients will be reviewed. Carboxylesterases may hydrolyze ester-based API, prodrugs and ester-bond containing polymer excipients, whereas lipases digest lipid formulation excipients, such as mono-, di- and triglycerides. To clarify the conditions that should be mimicked when designing in vitro studies, we briefly review the upper gastrointestinal physiology and provide new data on the inter-individual variability of enzyme activities in human intestinal fluids. Afterwards, the methodology for studying enzymatic hydrolysis of API, prodrugs, lipid and polymeric excipients, as well as the main results that have been obtained, are summarized. In vitro digestion models used to characterize lipid formulations are well described, but data about the hydrolysis of lipid excipients (including surfactants) has been scarce and contradictory. Data on API and prodrug hydrolysis by esterases is available; however, inconsistent use of enzyme types and concentrations limits structure-stability relationships. Hydrolysis of polymer excipients in the lumen has not been significantly explored, with only qualitative data available for cellulose derivates, polyesters, starches, etc. Harmonization of the methodology is required in order to curate larger enzymatic hydrolysis datasets, which will enable mechanistic understanding and theoretical prediction.
Collapse
Affiliation(s)
- Zahari Vinarov
- Department of Chemical and Pharmaceutical Engineering, Faculty of Chemistry and Pharmacy, Sofia University "St. Kl. Ohridski", 1164 Sofia, Bulgaria.
| | - Anette Müllertz
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark; Bioneer: FARMA, Department of Pharmacy, University of Copenhagen, Universitetsparken 4, 2100 Copenhagen, Denmark
| | - Hristina Mircheva
- Department of Chemical and Pharmaceutical Engineering, Faculty of Chemistry and Pharmacy, Sofia University "St. Kl. Ohridski", 1164 Sofia, Bulgaria
| | | | | | - Sharon Pradeep Kumar
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13, 8010 Graz, Austria
| | - Amrit Paudel
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13, 8010 Graz, Austria
| | | | - Patrick Augustijns
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
7
|
Lucic B, Franciscato DS, Nogueira HP, Gallucci L, Silveira Junior AT, Ismail AM, Robinson M, Dallinger T, Gutfleisch C, Kurz J, Toledo M, Dias da Silva Ferraz J, Tarek M, Dias D, Diaz RS, ElHefnawi M, Forcato M, Monteiro HP, Lusic M, Shytaj IL, Savarino A. Fast pH-Driven Solubilization Method of Realgar (As 4S 4) to Reduce the Toxicity of Arsenic [As(III)] for Medicinal Purposes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2502740. [PMID: 40271739 DOI: 10.1002/advs.202502740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Indexed: 04/25/2025]
Abstract
Acute promyelocytic leukemia (APL) accounts for 5-15% of acute myeloid leukemia cases. It is typically characterized by the (15;17) chromosomal translocation, producing the pathogenic retinoic acid receptor (RAR) alpha/promyelocytic leukemia (PML) fusion protein. Recently, remission of APL has been achieved using the first chemotherapy-independent oral drug regimen in anticancer therapy, consisting of all-trans retinoic acid (targeting RARalpha) and the arsenic sulfide realgar (targeting PML). However, clinical adoption of realgar and the characterization of its active breakdown products have been hampered by its poor solubility. Here, a scalable pH/temperature-based process is described that partially mimics gut transition, achieving fast and reproducible solubilization of realgar. Six different spectroscopic and spectrometric techniques are employed to investigate solubilized realgar. Furthermore, it is shown that solubilized realgar targets PML, displaying wider in vitro therapeutic indices and lower off-target effects than arsenic trioxide, the current APL standard of care. Moreover, in line with evidence of an interplay between PML and HIV persistence, solubilized realgar can disrupt HIV latency, the main barrier to an HIV/AIDS cure, in CD4 T cells of people living with HIV. These findings may open avenues for streamlining realgar solubilization and designing less toxic, orally administrable arsenic-based therapies.
Collapse
Affiliation(s)
- Bojana Lucic
- Department of Infectious Diseases, Integrative Virology, Heidelberg University, 69120, Heidelberg, Germany
- German Center for Infection Research, 69120, Heidelberg, Germany
| | | | | | - Lara Gallucci
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | | | - Asmaa Mohamed Ismail
- Spectroscopy Department, National Research Centre, 33 El Bohouth Street, Dokki, Giza, 12622, Egypt
| | - Millie Robinson
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | - Teresa Dallinger
- Department of Infectious Diseases, Integrative Virology, Heidelberg University, 69120, Heidelberg, Germany
| | - Claudia Gutfleisch
- Center for Infectious Diseases, Medical Microbiology und Hygiene, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Jochen Kurz
- Center for Infectious Diseases, Medical Microbiology und Hygiene, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Maytê Toledo
- Department of Biochemistry, Center for Cellular and Molecular Therapy, Federal University of São Paulo, São Paulo, 04021-001, Brazil
| | | | - Mohammad Tarek
- Clinical Hematology Department, Armed Forces College of Medicine (AFCM) Cairo Governatorate, Heliopolis, 11774, Egypt
| | - Danilo Dias
- Infectious Diseases Department, Federal University of São Paulo, São Paulo, 04021-001, Brazil
| | - Ricardo Sobhie Diaz
- Infectious Diseases Department, Federal University of São Paulo, São Paulo, 04021-001, Brazil
| | - Mahmoud ElHefnawi
- Informatics and Systems Department, National Research Centre, 33 El Bohouth Street, Dokki, Giza, 12622, Egypt
| | - Mattia Forcato
- Department of Molecular Medicine, University of Padova, Padova, 35122, Italy
| | - Hugo P Monteiro
- Department of Biochemistry, Center for Cellular and Molecular Therapy, Federal University of São Paulo, São Paulo, 04021-001, Brazil
| | - Marina Lusic
- Department of Infectious Diseases, Integrative Virology, Heidelberg University, 69120, Heidelberg, Germany
- German Center for Infection Research, 69120, Heidelberg, Germany
| | - Iart Luca Shytaj
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
- Infectious Diseases Department, Federal University of São Paulo, São Paulo, 04021-001, Brazil
| | - Andrea Savarino
- Department of Infectious Diseases, Italian Institute of Health, Rome, 00161, Italy
| |
Collapse
|
8
|
Ding S, Alexander E, Liang H, Kulchar RJ, Singh R, Herzog RW, Daniell H, Leong KW. Synthetic and Biogenic Materials for Oral Delivery of Biologics: From Bench to Bedside. Chem Rev 2025; 125:4009-4068. [PMID: 40168474 DOI: 10.1021/acs.chemrev.4c00482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
The development of nucleic acid and protein drugs for oral delivery has lagged behind their production for conventional nonoral routes. Over the past decade, the evolution of DNA- and RNA-based technologies combined with the innovation of state-of-the-art delivery vehicles for nucleic acids has brought rapid advancements to the biopharmaceutical field. Nucleic acid therapies have the potential to achieve long-lasting effects, or even cures, by inhibiting or editing genes, which is not possible with conventional small-molecule drugs. However, challenges and limitations must be addressed before these therapies can provide cures for chronic conditions and rare diseases, rather than only offering temporary relief. Nucleic acids and proteins face premature degradation in the acidic, enzyme-rich stomach environment and are rapidly cleared by the liver. To overcome these challenges, various delivery vehicles have been developed to transport therapeutic compounds to the intestines, where the active compounds are released and gut microbiota and mucosal immune system also play an important role. This review provides a comprehensive overview of the promises and pitfalls associated with the oral route of administration of biologics, current delivery systems, applications of orally delivered therapeutics, and the challenges and considerations for translation of nucleic acid and protein therapeutics into clinical practice.
Collapse
Affiliation(s)
- Suwan Ding
- Department of Biomedical Engineering, Columbia University, 500 West 120th Street, New York, New York 10027, United States
| | - Elena Alexander
- Department of Biomedical Engineering, Columbia University, 500 West 120th Street, New York, New York 10027, United States
| | - Huiyi Liang
- Department of Biomedical Engineering, Columbia University, 500 West 120th Street, New York, New York 10027, United States
| | - Rachel J Kulchar
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, 240 South 40th Street, Philadelphia, Pennsylvania 19104, United States
| | - Rahul Singh
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, 240 South 40th Street, Philadelphia, Pennsylvania 19104, United States
| | - Roland W Herzog
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Henry Daniell
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, 240 South 40th Street, Philadelphia, Pennsylvania 19104, United States
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, 500 West 120th Street, New York, New York 10027, United States
| |
Collapse
|
9
|
Subramanian D, Chin A, Shi Y, Liu GW, Langer R, Traverso G. Identification and Validation of Cyclic Peptides with Mucin-Selective, Location-Specific Binding in the Gastrointestinal Tract. ACS NANO 2025; 19:14693-14706. [PMID: 40216380 PMCID: PMC12020424 DOI: 10.1021/acsnano.4c13520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 03/22/2025] [Accepted: 03/24/2025] [Indexed: 04/23/2025]
Abstract
Oral drug delivery is a widely preferred method of drug administration due to its ease of use and convenience for patients. Localization of drug release in the gastrointestinal (GI) tract is important to treat localized diseases and maximize drug absorption. However, achieving drug localization in the dynamic GI tract is challenging. To address this challenge, we leveraged the geographic diversity of the GI tract by targeting its mucus layers, which coat the epithelial surfaces. These layers, composed of mucin glycoproteins, are synthesized with unique chemical compositions and expressed in different regions, making them ideal targets for drug localization. In this article, we identify cyclic peptides that bind selectively to MUC2 (in the intestines) and MUC5AC (in the stomach), serving as targeting ligands to these regions of the GI tract. We demonstrate the effectiveness of these peptides through in vitro, ex vivo, and in vivo experiments, showing that incorporating these targeting ligands can increase binding and selectivity 2-fold to the desired regions, thus potentially overcoming challenges with localizing drug distribution in oral delivery. These results indicate that cyclic peptides can be used to localize drug cargoes at certain sites in the body compared to free drugs.
Collapse
Affiliation(s)
- Deepak
A. Subramanian
- Department
of Chemical Engineering, Massachusetts Institute
of Technology, Cambridge, Massachusetts 02139, United States
- David
H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Austin Chin
- Department
of Chemical Engineering, Massachusetts Institute
of Technology, Cambridge, Massachusetts 02139, United States
| | - Yunhua Shi
- David
H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Gary W. Liu
- David
H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Robert Langer
- Department
of Chemical Engineering, Massachusetts Institute
of Technology, Cambridge, Massachusetts 02139, United States
- David
H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department
of Mechanical Engineering, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Giovanni Traverso
- David
H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department
of Mechanical Engineering, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
- Division
of Gastroenterology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
10
|
Kozu H, Kobayashi I, Ichikawa S. A Review on In Vitro Evaluation of Chemical and Physical Digestion for Controlling Gastric Digestion of Food. Foods 2025; 14:1435. [PMID: 40282836 PMCID: PMC12027026 DOI: 10.3390/foods14081435] [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: 03/12/2025] [Revised: 04/07/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025] Open
Abstract
Food digestion in the gastrointestinal is a series of processes consisting of chemical and physical digestion. Recently, developing foods with controlled digestion in the stomach may attract more attention. Hydrogel foods are useful tools for designing foods with controlled digestion because it is relatively easy to design their food characteristics by adjusting the type and content of the additives. This review introduces the latest status of in vitro gastric digestion as a food characterization system. The in vitro evaluation of chemical gastric digestion by gastric acid and digestive enzymes focuses on INFOGEST-standardized gastrointestinal digestion protocols for healthy adults, infants, and older adults. For the in vitro evaluation of physical gastric digestion by peristalsis, the current development of gastrointestinal tract devices that precisely or efficiently simulate the shape of the stomach and gastric peristalsis is described. In addition, we introduce studies that have utilized these devices to investigate the gastric digestion behavior of hydrocolloid foods with different mechanical characteristics.
Collapse
Affiliation(s)
- Hiroyuki Kozu
- Institute of Food Research, NARO, 2-1-12 Kannondai, Tsukuba 305–8642, Ibaraki, Japan;
| | - Isao Kobayashi
- Institute of Food Research, NARO, 2-1-12 Kannondai, Tsukuba 305–8642, Ibaraki, Japan;
| | - Sosaku Ichikawa
- Institute of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305–8572, Ibaraki, Japan
| |
Collapse
|
11
|
Tronel A, Roger-Margueritat M, Plazy C, Biennier S, Craspay A, Mohanty I, Portier SC, Laiola M, Roeselers G, Mathieu N, Hupe M, Dorrestein PC, Alcaraz JP, Martin D, Cinquin P, Silvent AS, Giai J, Proust M, Soranzo T, Buelow E, Gouellec ALE. Profiling the human luminal small intestinal microbiome using a novel ingestible medical device. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.18.25326056. [PMID: 40321269 PMCID: PMC12047917 DOI: 10.1101/2025.04.18.25326056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
The invasive nature of sample collection for studying the small intestinal (SI) microbiome often results in its poor characterization. This study evaluated a novel ingestible medical device (MD) for SI luminal sample collection. A monocentric interventional trial (NCT05477069) was conducted on 15 healthy subjects. Metagenomics, metabolomics and culturomics assessed the MD's effectiveness in characterizing the healthy SI microbiome and identifying potential biomarkers. The SI microbiota differed significantly from the fecal microbiota, displaying high inter-individual variability, lower species richness, and reduced alpha diversity. A combined untargeted and semi-targeted LC-MS/MS metabolomics approach identified a distinct SI metabolic footprint, with bile acids and amino acids being the most abundant classes of metabolites. Host and host/microbe-derived bile acids were particularly abundant in SI samples. The application of a fast culturomics approach to two SI samples enabled species-level characterization, resulting in the identification of 90 bacterial species, including five potential novel species. The present study demonstrates the efficacy of our novel sampling MD in enabling comprehensive SI microbiome analysis through an integrative multi-omics approach, allowing the identification of distinct microbiome signatures between SI and fecal samples.
Collapse
Affiliation(s)
- Alexandre Tronel
- Pelican Health, 5 avenue du Grand Sablon, 38700, La Tronche, France
- Univ. Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, CHU Grenoble Alpes, TIMC, 38000 Grenoble, France
| | - Morgane Roger-Margueritat
- Univ. Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, CHU Grenoble Alpes, TIMC, 38000 Grenoble, France
| | - Caroline Plazy
- Univ. Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, CHU Grenoble Alpes, TIMC, 38000 Grenoble, France
- Service de Biochimie Biologie Moléculaire Toxicologie Environnementale, UM Biochimie des Enzymes et des Protéines, Institut de Biologie et Pathologie, CHU Grenoble-Alpes, 38000 Grenoble, France
- Plateforme de Métabolomique GEMELI-GExiM, Institut de Biologie et Pathologie, CHU Grenoble-Alpes, 38000 Grenoble, France
| | - Salomé Biennier
- Pelican Health, 5 avenue du Grand Sablon, 38700, La Tronche, France
| | - Anthony Craspay
- Pelican Health, 5 avenue du Grand Sablon, 38700, La Tronche, France
| | - Ipsita Mohanty
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Stéphanie Cools Portier
- Danone Global Research & Innovation Center, Route départementale 128, 91 190 Gif sur Yvette, France
| | - Manolo Laiola
- Danone Global Research & Innovation Center, Route départementale 128, 91 190 Gif sur Yvette, France
| | - Guus Roeselers
- Danone Global Research & Innovation Center, Route départementale 128, 91 190 Gif sur Yvette, France
| | - Nicolas Mathieu
- Univ. Grenoble Alpes/Hepato-Gastroenterology and Digestive Oncology Department, CHU Grenoble Alpes/Institute for Advanced Biosciences, CNRS UMR 5309-INSERM U1209, 38043 Grenoble, France
| | - Marianne Hupe
- Univ. Grenoble Alpes/Hepato-Gastroenterology and Digestive Oncology Department, CHU Grenoble Alpes/Institute for Advanced Biosciences, CNRS UMR 5309-INSERM U1209, 38043 Grenoble, France
| | - Pieter C Dorrestein
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Jean-Pierre Alcaraz
- Univ. Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, CHU Grenoble Alpes, TIMC, 38000 Grenoble, France
| | - Donald Martin
- Univ. Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, CHU Grenoble Alpes, TIMC, 38000 Grenoble, France
| | - Philippe Cinquin
- Univ. Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, CHU Grenoble Alpes, TIMC, 38000 Grenoble, France
| | - Anne-Sophie Silvent
- Univ. Grenoble Alpes, Inserm, CHU Grenoble Alpes, CIC, 38000 Grenoble, France
| | - Joris Giai
- Univ. Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, CHU Grenoble Alpes, TIMC, 38000 Grenoble, France
- Univ. Grenoble Alpes, Inserm, CHU Grenoble Alpes, CIC, 38000 Grenoble, France
| | - Marion Proust
- Univ. Grenoble Alpes, Inserm, CHU Grenoble Alpes, CIC, 38000 Grenoble, France
| | - Thomas Soranzo
- Pelican Health, 5 avenue du Grand Sablon, 38700, La Tronche, France
| | - Elena Buelow
- Univ. Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, CHU Grenoble Alpes, TIMC, 38000 Grenoble, France
| | - Audrey LE Gouellec
- Univ. Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, CHU Grenoble Alpes, TIMC, 38000 Grenoble, France
- Service de Biochimie Biologie Moléculaire Toxicologie Environnementale, UM Biochimie des Enzymes et des Protéines, Institut de Biologie et Pathologie, CHU Grenoble-Alpes, 38000 Grenoble, France
- Plateforme de Métabolomique GEMELI-GExiM, Institut de Biologie et Pathologie, CHU Grenoble-Alpes, 38000 Grenoble, France
| |
Collapse
|
12
|
Blaesi AH, Saka N. Gastroretentive fibrous dosage forms for prolonged delivery of sparingly-soluble tyrosine kinase inhibitors. Part 3: Theoretical models of drug concentration in blood. Int J Pharm 2025; 674:124362. [PMID: 38901538 DOI: 10.1016/j.ijpharm.2024.124362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/12/2024] [Accepted: 06/16/2024] [Indexed: 06/22/2024]
Abstract
In this part, drug concentration in blood after ingesting gastroretentive fibrous and immediate-release particulate dosage forms is modeled. The tyrosine kinase inhibitor nilotinib, which is slightly soluble in low-pH gastric fluid but practically insoluble in pH-neutral intestinal fluid is used as drug. The models suggest that upon ingestion, the fibrous dosage form expands, is retained in the stomach for prolonged time, and releases drug into the gastric fluid at a constant rate. The released drug molecules flow into the duodenum with the gastric fluid, and are absorbed by the blood. The drug is eliminated from the blood by the liver at a rate proportional to its concentration. Thus, as the drug concentration in blood increases due to absorption, the elimination rate increases, too, and will eventually be the same as the absorption rate, so that the drug concentration in blood plateaus out. After the gastric residence time, drug absorption stops, and the drug concentration in blood drops to zero. By contrast, after administering the immediate-release particulate dosage form the drug particles are swept out of the stomach rapidly, and drug absorption stops much earlier. The drug concentration in blood rises and falls without attaining steady state. The gastroretentive fibrous dosage forms enable a constant drug concentration in blood for drugs that are insoluble in intestinal fluids.
Collapse
Affiliation(s)
- Aron H Blaesi
- Enzian Pharmaceutics Blaesi AG, CH-7078 Lenzerheide, Switzerland; Enzian Pharmaceutics, Inc., Cambridge, MA 02139, USA.
| | - Nannaji Saka
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
13
|
Chen P, Wang Y, Cai Z, Lu X. Enhanced bioaccessibility of cyclolinopeptides via zein-cyclodextrin nanoparticles: Simulated gastrointestinal digestion and cellular uptake study. Food Chem 2025; 471:142841. [PMID: 39808983 DOI: 10.1016/j.foodchem.2025.142841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 12/28/2024] [Accepted: 01/08/2025] [Indexed: 01/16/2025]
Abstract
Cyclolinopeptides (CLS) are hydrophobic cyclic peptides in flaxseed with multiple bioactive activities. This study developed zein (Z)-cyclodextrin (CD) binary nanoparticles (NPs) as an oral delivery system for CLS. Z-CD NP had a smaller diameter (Dh) and better encapsulation effect on CLS. Formation of CLS-loaded NPs was driven by hydrogen bonds and electrostatic interactions. Presence of CD improved the thermal, pH and storage stabilities of NPs. Besides, CD prevented premature release of CLS in the stomach and enhanced the bioaccessibility of CLS to a maximum of 86.71 % ± 2.20 %. Lipid-raft-mediated endocytosis was involved in the cell uptake of NPs, where the addition of CD significantly facilitated the uptake of NPs. Z-CD NPs also enhanced absorption and reduced secretion of CLS after digestion. Overall, this study provides a simple approach to enhance the oral delivery efficiency of CLS by modulating Z-based NPs with CD.
Collapse
Affiliation(s)
- Peifang Chen
- Department of Food Science and Engineering, Jinan University, Guangzhou 510632, China
| | - Yong Wang
- Department of Food Science and Engineering, Jinan University, Guangzhou 510632, China; Guangdong Engineering Technology Research Center for Cereal and Oil Byproduct Biorefinery, Guangzhou 510632, China; Guangdong Joint International Centre of Oilseed Biorefinery, Nutrition and Safety, Guangzhou 510632, China.
| | - Zizhe Cai
- Department of Food Science and Engineering, Jinan University, Guangzhou 510632, China
| | - Xuanxuan Lu
- Department of Food Science and Engineering, Jinan University, Guangzhou 510632, China; Guangdong Engineering Technology Research Center for Cereal and Oil Byproduct Biorefinery, Guangzhou 510632, China; Guangdong Joint International Centre of Oilseed Biorefinery, Nutrition and Safety, Guangzhou 510632, China.
| |
Collapse
|
14
|
Subramanian DA, Kirtane AR, White GN, Freitas DE, Ishida K, Jenkins J, Pettinari A, Morimoto J, Fitzgerald N, Traverso G. Identification and Validation of Small Molecules with Mucin-Selective Regiospecific Binding in the Gastrointestinal Tract. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.31.646052. [PMID: 40235971 PMCID: PMC11996415 DOI: 10.1101/2025.03.31.646052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Oral drug delivery is a widely used method of drug administration; however, achieving localized drug release at specific regions of the gastrointestinal (GI) tract is generally accomplished by using broad environmental differences. The GI tract is a complex system with regional differences in composition, such as selective expression of mucin glycoproteins in different organs. Here, we identify small molecule ligands that can selectively bind to the different mucins to localize drug delivery to the small intestine and stomach. We demonstrate up to a 10-fold increase in particle binding to these organs and up to a 4-fold increase in selectivity compared to chitosan. Additionally, we observe up to a 9-fold increase in budesonide concentration in the small intestine and a 25-fold increase in tetracycline concentration in the stomach. These results show that we have developed a versatile platform capable of sequestering a variety of drugs in certain GI tract organs.
Collapse
|
15
|
Ben-Haim AE, Shalev N, Amalraj AJJ, Zelinger E, Mani KA, Belausov E, Shoval I, Nativ-Roth E, Maria R, Atkins A, Sadashiva R, Koltai H, Mechrez G. Nanocarriers for cancer-targeted delivery based on Pickering emulsions stabilized by casein nanoparticles. Int J Biol Macromol 2025; 298:140822. [PMID: 39929470 DOI: 10.1016/j.ijbiomac.2025.140822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 02/03/2025] [Accepted: 02/07/2025] [Indexed: 02/26/2025]
Abstract
This study demonstrates the development of stimuli-responsive Pickering emulsions stabilized by casein nanoparticles (CNPs) for targeted drug delivery to colorectal cancer cells (CRC). Encapsulation of a fluorescent dye simulates therapeutic delivery, demonstrating biomedical potential. The oil-in-water nanoemulsions stabilized by CNPs function as nanocarriers sensitive to matrix metalloproteinase-7 (MMP-7), an enzyme overexpressed in CRC cells, enabling precise drug release. Emulsions exhibited strong stability due CNPs forming a robust layer at the oil-water interface, enhancing bioavailability and controlled release. Covalent modifications of CNPs with polyethyleneimine (PEI) or polyacrylic acid (PAA), and pH adjustments optimize the zeta potential, improving surface charge and delivery efficiency. Maximal CNP uptake occurred with PAA-modified CNPs (-20 mV), showing superior interaction with CRC cells compared to pristine (-6.7 mV) and PEI-modified (+30.5, +42.1 mV) CNPs. Confocal microscopy and imaging flow cytometry confirmed that CNP-stabilized emulsions enhance CRC inter-localization compared to dispersed CNPs. Nanoemulsions with the highest CNP uptake showed selective interaction with tumor cells, while minimizing oil droplet uptake, driven by nanoscale dimensions and targeted surface interactions. Enzymatic degradation of CNPs by MMP-7 induces phase separation and targeted release. This dual-functional system, leveraging charge modification and enzymatic responsiveness, highlights CNP-stabilized nanoemulsions as a promising CRC-targeted drug delivery platform.
Collapse
Affiliation(s)
- Avital Ella Ben-Haim
- Department of Food Sciences, Institute of Postharvest and Food Sciences, Agricultural Research Organization (ARO), Volcani Institute, 68 HaMaccabim Road, Rishon Lezion 7505101, Israel; The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, POB 12, Rehovot 7610001, Israel
| | - Nurit Shalev
- Institute of Plant Sciences, Agricultural Research Organization (ARO), The Volcani Institute, PO Box 6, Beit Dagan 50250, Israel
| | - Antolin Jesila Jesu Amalraj
- Department of Food Sciences, Institute of Postharvest and Food Sciences, Agricultural Research Organization (ARO), Volcani Institute, 68 HaMaccabim Road, Rishon Lezion 7505101, Israel
| | - Einat Zelinger
- The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, POB 12, Rehovot 7610001, Israel
| | - Karthik Ananth Mani
- Department of Food Sciences, Institute of Postharvest and Food Sciences, Agricultural Research Organization (ARO), Volcani Institute, 68 HaMaccabim Road, Rishon Lezion 7505101, Israel; The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, POB 12, Rehovot 7610001, Israel
| | - Eduard Belausov
- Department of Ornamental Plants and Agricultural Biotechnology, Institute of Plant Sciences, Agricultural Research Organization (ARO), Volcani Institute, 68 HaMaccabim Road, Rishon Lezion 7505101, Israel
| | - Irit Shoval
- The Kanbar core facility unit, The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Israel
| | - Einat Nativ-Roth
- Ilse Katz Institute for Nanoscale Science and Technology, Ben Gurion University of the Negev, Israel
| | - Raquel Maria
- Ilse Katz Institute for Nanoscale Science and Technology, Ben Gurion University of the Negev, Israel
| | - Ayelet Atkins
- Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan 52900, Israel
| | - Rajitha Sadashiva
- Institute of Plant Sciences, Agricultural Research Organization (ARO), The Volcani Institute, PO Box 6, Beit Dagan 50250, Israel
| | - Hinanit Koltai
- Institute of Plant Sciences, Agricultural Research Organization (ARO), The Volcani Institute, PO Box 6, Beit Dagan 50250, Israel
| | - Guy Mechrez
- Department of Food Sciences, Institute of Postharvest and Food Sciences, Agricultural Research Organization (ARO), Volcani Institute, 68 HaMaccabim Road, Rishon Lezion 7505101, Israel.
| |
Collapse
|
16
|
Shao H, Liu M, Jiang H, Zhang Y. Polysaccharide-based drug delivery targeted approach for colon cancer treatment: A comprehensive review. Int J Biol Macromol 2025; 302:139177. [PMID: 39798740 DOI: 10.1016/j.ijbiomac.2024.139177] [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: 08/28/2024] [Revised: 12/10/2024] [Accepted: 12/23/2024] [Indexed: 01/15/2025]
Abstract
Colon cancer is a leading cause of cancer-related morbidity and mortality worldwide, necessitating advancements in therapeutic strategies to improve outcomes. Current treatment modalities, including surgery, chemotherapy, and radiation, are limited by systemic toxicity, low drug utilization rates, and off-target effects. Colon-targeted drug delivery systems (CDDS) offer a promising alternative by leveraging the colon's unique physiology, such as near-neutral pH and extended transit time, to achieve localized and controlled drug release. Polysaccharide-based CDDS, utilizing natural polymers like chitosan, cyclodextrin, pectin, guar gum, alginate, hyaluronic acid, dextran, chondroitin sulfate, and inulin, have emerged as innovative approaches for improving the specificity and efficacy of colon cancer treatments. These biocompatible and biodegradable polymers enable site-specific drug delivery, enhance tumor apoptosis, reduce systemic side effects, and improve patient compliance. This review evaluates recent advancements in polysaccharide-based CDDS, detailing their drug release mechanisms, therapeutic potential, and challenges in overcoming gastrointestinal transit and pH variability. Studies highlight the successful formulation of nanoparticles, microspheres, and other delivery systems, demonstrating targeted drug delivery, improved bioavailability, and enhanced cytotoxicity against colon cancer cells in-vitro and in-vivo. The review underscores the need for continued research on polysaccharide-based CDDS for colon cancer treatment, offering a path toward more effective, patient-centered oncological care.
Collapse
Affiliation(s)
- Hua Shao
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning, China
| | - Minghua Liu
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning, China
| | - Hongfang Jiang
- Department of Geriatrics, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning, China.
| | - Ying Zhang
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
17
|
Walter S, Schatz V, Petzold J, Schmidt C, Hoffmann S, Jantsch J, Gerlach RG. O 2-dependent incapacitation of the Salmonella pathogenicity island 1 repressor HilE. Front Cell Infect Microbiol 2025; 15:1434254. [PMID: 40041146 PMCID: PMC11876186 DOI: 10.3389/fcimb.2025.1434254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 01/27/2025] [Indexed: 03/06/2025] Open
Abstract
For successful colonization, pathogenic bacteria need to adapt their metabolism and virulence functions to challenging environments within their mammalian hosts that are frequently characterized by low oxygen (O2) tensions. Upon oral ingestion, the human pathogen Salmonella enterica serovar Typhimurium (S. Typhimurium) is exposed to changing O2 and pH levels. Low concentrations of O2, which can enhance the virulence of enteroinvasive pathogens, facilitate the expression of the type three secretion system (T3SS-1) encoded by the Salmonella pathogenicity island 1 (SPI-1) that is critical for enteroinvasion and pathogenicity of S. Typhimurium. To study the impact of key environmental cues of the intestine when Salmonella encounter enterocytes, we established an in vitro growth model, which allows shifting the concentration of O2 from 0.5% to 11% and the pH from 5.9 to 7.4 in the presence of acetate and the alternative electron acceptor nitrate. Compared to normoxia, hypoxia elevated the expression of SPI-1 genes encoding T3SS-1 translocators and effectors, which resulted in higher invasion and effector translocation in epithelial cells. While hypoxia and pH shift only marginally altered the gene expression of SPI-1 regulators, including the SPI-1 repressor hilE, hypoxia and pH shift completely incapacitated HilE in a post-translational manner, ultimately promoting SPI-1 activity. From these findings, we conclude that O2-dependent HilE function allows for ultrasensitive adaptation of SPI-1 activity in environments with varying O2 availability such as the intestinal tract.
Collapse
Affiliation(s)
- Steffi Walter
- Project Group 5, Robert Koch Institute, Wernigerode, Germany
| | - Valentin Schatz
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg and University of Regensburg, Regensburg, Germany
- Institute for Medical Microbiology, Immunology, and Hygiene, University Hospital Cologne and Faculty of Medicine, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Jana Petzold
- Mikrobiologisches Institut – Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | | | | | - Jonathan Jantsch
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg and University of Regensburg, Regensburg, Germany
- Institute for Medical Microbiology, Immunology, and Hygiene, University Hospital Cologne and Faculty of Medicine, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Roman G. Gerlach
- Project Group 5, Robert Koch Institute, Wernigerode, Germany
- Mikrobiologisches Institut – Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
18
|
Dmytriv TR, Lushchak O, Lushchak VI. Glucoraphanin conversion into sulforaphane and related compounds by gut microbiota. Front Physiol 2025; 16:1497566. [PMID: 39995480 PMCID: PMC11847849 DOI: 10.3389/fphys.2025.1497566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 01/22/2025] [Indexed: 02/26/2025] Open
Abstract
Glucosinolate glucoraphanin, common in cruciferous vegetables, is a biologically stable precursor of isothiocyanates, such as sulforaphane and erucin, potent activators of Nrf2 signaling coordinating an adaptive response to oxidative stress. Sulforaphane is formed by the hydrolysis of glucoraphanin by a plant enzyme called myrosinase, which is inactivated in the stomach of mammals. Since the latter do not have enzymes possessing myrosinase-like activity, glucoraphanin can be metabolized by the gut microbiota, to sulforaphane, sulforaphane-nitrile, glucoerucin, erucin, and erucin-nitrile. Emerging evidence suggests that variations in gut microbiota composition significantly influence the efficiency and outcome of glucoraphanin metabolism, while sulforaphane itself may reciprocally modulate gut microbiota composition and functionality. This review examines the bidirectional interactions between glucoraphanin, sulforaphane, and gut microbiota. We assume that sulforaphane alleviates intestinal inflammation and oxidative stress maintaining intestinal homeostasis and gut barrier integrity. Besides, the role of sulforaphane in breaking the vicious cycle of oxidative stress and gut dysbiosis is reported, demonstrating the potential of dietary isothiocyanates to support gut barrier function.
Collapse
Affiliation(s)
- Tetiana R. Dmytriv
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
- Research and Development University, Ivano-Frankivsk, Ukraine
| | - Oleh Lushchak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
- Research and Development University, Ivano-Frankivsk, Ukraine
| | - Volodymyr I. Lushchak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
- Research and Development University, Ivano-Frankivsk, Ukraine
| |
Collapse
|
19
|
Salvador-Erro J, Pastor Y, Gamazo C. Targeting Enterotoxins: Advancing Vaccine Development for Enterotoxigenic Escherichia coli ETEC. Toxins (Basel) 2025; 17:71. [PMID: 39998088 PMCID: PMC11860656 DOI: 10.3390/toxins17020071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/29/2025] [Accepted: 02/04/2025] [Indexed: 02/26/2025] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is a major cause of diarrheal disease worldwide, particularly in children in low- and middle-income countries. Its ability to rapidly colonize the intestinal tract through diverse colonization factors and toxins underpins its significant public health impact. Despite extensive research and several vaccine candidates reaching clinical trials, no licensed vaccine exists for ETEC. This review explores the temporal and spatial coordination of ETEC virulence factors, focusing on the interplay between adherence mechanisms and toxin production as critical targets for therapeutic intervention. Advancements in molecular biology and host-pathogen interaction studies have uncovered species-specific variations and cross-reactivity between human and animal strains. In particular, the heat-labile (LT) and heat-stable (ST) toxins have provided crucial insights into molecular mechanisms and intestinal disruption. Additional exotoxins, such as EAST-1 and hemolysins, further highlight the multifactorial nature of ETEC pathogenicity. Innovative vaccine strategies, including multiepitope fusion antigens (MEFAs), mRNA-based approaches, and glycoconjugates, aim to enhance broad-spectrum immunity. Novel delivery methods, like intradermal immunization, show promise in eliciting robust immune responses. Successful vaccination against ETEC will offer an effective and affordable solution with the potential to greatly reduce mortality and prevent stunting, representing a highly impactful and cost-efficient solution to a critical global health challenge.
Collapse
Affiliation(s)
| | | | - Carlos Gamazo
- Department of Microbiology and Parasitology, Navarra Medical Research Institute (IdiSNA), University of Navarra, 31008 Pamplona, Spain; (J.S.-E.); (Y.P.)
| |
Collapse
|
20
|
Zhang W, Jia J, Yang Y, Ye D, Li Y, Li D, Wang J. Estradiol metabolism by gut microbiota in women's depression pathogenesis: inspiration from nature. Front Psychiatry 2025; 16:1505991. [PMID: 39935532 PMCID: PMC11811108 DOI: 10.3389/fpsyt.2025.1505991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 01/10/2025] [Indexed: 02/13/2025] Open
Abstract
The recurrence and treatment resistance of depression remain significant issues, primarily due to an inadequate understanding of its pathogenesis. Recent scientific evidence indicates that gut microbiota influence estradiol metabolism and are associated with the development of depression in nonpremenopausal women. Integrating existing studies on the regulation of estradiol metabolism by microorganisms in nature and the relevance of its degradation products to depression, recent scientific explorations have further elucidated the key mechanisms by which gut microbiota catabolize estradiol through specific metabolic pathways. These emerging scientific findings suggest that the unique metabolic effects of gut microbiota on estradiol may be one of the central drivers in the onset and course of depression in non-menopausal women.
Collapse
Affiliation(s)
- Wei Zhang
- Division of Colorectal Surgery, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan, China
| | - Jinghan Jia
- Division of Colorectal Surgery, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan, China
| | - Yuhang Yang
- Division of Colorectal Surgery, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan, China
| | - Dawei Ye
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yan Li
- Neurology, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan, China
| | - Di Li
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinxi Wang
- Division of Colorectal Surgery, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
21
|
Sun H, Xue X, Robilotto GL, Zhang X, Son C, Chen X, Cao Y, Nan K, Yang Y, Fennell G, Jung J, Song Y, Li H, Lu SH, Liu Y, Li Y, Zhang W, He J, Wang X, Li Y, Mickle AD, Zhang Y. Liquid-based encapsulation for implantable bioelectronics across broad pH environments. Nat Commun 2025; 16:1019. [PMID: 39863617 PMCID: PMC11762702 DOI: 10.1038/s41467-025-55992-x] [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: 07/06/2024] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
Wearable and implantable bioelectronics that can interface for extended periods with highly mobile organs and tissues across a broad pH range would be useful for various applications in basic biomedical research and clinical medicine. The encapsulation of these systems, however, presents a major challenge, as such devices require superior barrier performance against water and ion penetration in challenging pH environments while also maintaining flexibility and stretchability to match the physical properties of the surrounding tissue. Current encapsulation materials are often limited to near-neutral pH conditions, restricting their application range. In this work, we report a liquid-based encapsulation approach for bioelectronics under extreme pH environments. This approach achieves high optical transparency, stretchability, and mechanical durability. When applied to implantable wireless optoelectronic devices, our encapsulation method demonstrates outstanding water resistance in vitro, ranging from extremely acidic environments (pH = 1.5 and 4.5) to alkaline conditions (pH = 9). We also demonstrate the in vivo biocompatibility of our encapsulation approach and show that encapsulated wireless optoelectronics maintain robust operation throughout 3 months of implantation in freely moving mice. These results indicate that our encapsulation strategy has the potential to protect implantable bioelectronic devices in a wide range of research and clinical applications.
Collapse
Affiliation(s)
- He Sun
- Department of Biomedical Engineering and the Institute of Materials Science, University of Connecticut, Storrs, CT, 06269, USA
| | - Xiaoting Xue
- Department of Biomedical Engineering and the Institute of Materials Science, University of Connecticut, Storrs, CT, 06269, USA.
| | - Gabriella L Robilotto
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, 32603, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Xincheng Zhang
- Department of Biomedical Engineering and the Institute of Materials Science, University of Connecticut, Storrs, CT, 06269, USA
| | - ChangHee Son
- Department of Biomedical Engineering and the Institute of Materials Science, University of Connecticut, Storrs, CT, 06269, USA
| | - Xingchi Chen
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, 32306, USA
| | - Yue Cao
- Department of Biomedical Engineering and the Institute of Materials Science, University of Connecticut, Storrs, CT, 06269, USA
| | - Kewang Nan
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Yiyuan Yang
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Gavin Fennell
- Department of Biomedical Engineering and the Institute of Materials Science, University of Connecticut, Storrs, CT, 06269, USA
| | - Jaewook Jung
- Department of Biomedical Engineering and the Institute of Materials Science, University of Connecticut, Storrs, CT, 06269, USA
| | - Yang Song
- Department of Biomedical Engineering and the Institute of Materials Science, University of Connecticut, Storrs, CT, 06269, USA
| | - Huijie Li
- Department of Biomedical Engineering and the Institute of Materials Science, University of Connecticut, Storrs, CT, 06269, USA
| | - Shao-Hao Lu
- Department of Materials Science and Engineering and the Institute of Materials Science, University of Connecticut, Storrs, CT, 06269, USA
| | - Yizhou Liu
- Department of Biomedical Engineering and the Institute of Materials Science, University of Connecticut, Storrs, CT, 06269, USA
| | - Yi Li
- Department of Materials Science and Engineering and the Institute of Materials Science, University of Connecticut, Storrs, CT, 06269, USA
| | - Weiyi Zhang
- Department of Biomedical Engineering and the Institute of Materials Science, University of Connecticut, Storrs, CT, 06269, USA
| | - Jie He
- Department of Chemistry and the Institute of Materials Science, University of Connecticut, Storrs, CT, 06269, USA
| | - Xueju Wang
- Department of Materials Science and Engineering and the Institute of Materials Science, University of Connecticut, Storrs, CT, 06269, USA
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, 32306, USA
| | - Aaron D Mickle
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, 32603, USA.
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
- J. Crayton Pruitt Family Department of Biomedical Engineering, College of Engineering, University of Florida, Gainesville, FL, 32610, USA.
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
- Department of Urology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
| | - Yi Zhang
- Department of Biomedical Engineering and the Institute of Materials Science, University of Connecticut, Storrs, CT, 06269, USA.
| |
Collapse
|
22
|
Koshani R, Yeh SL, He Z, Narasimhalu N, Vom Steeg LG, Sim DG, Woods RJ, Read AF, Sheikhi A. Polymeric Anti-Antibiotic Microparticles to Prevent Antibiotic Resistance Evolution. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2407549. [PMID: 39828608 DOI: 10.1002/smll.202407549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 11/23/2024] [Indexed: 01/22/2025]
Abstract
Vancomycin (VAN) and daptomycin (DAP) are among the last-resort antibiotics for treating multidrug-resistant Gram-positive bacterial infections. They are administered intravenously (IV); however, ≈5 - 10% of the total IV dose is released in the gastrointestinal (GI) tract via biliary excretion, driving resistance emergence in commensal Enterococcus faecium (E. faecium) populations. Here, it is reported that sevelamer (SEV), a Food and Drug Administration (FDA)-approved anion-exchange polymeric microparticle, captures anionic DAP within minutes and cationic VAN within hours, inactivating the antibacterial efficacy of DAP and VAN. In vitro SEV-mediated VAN or DAP transient removal is successfully described by a diffusion-adsorption mechanism. In vivo oral SEV treatment effectively prevented VAN resistance enrichment following the VAN treatment of E. faecium-colonized mice. This work shows, for the first time, that the adjuvant SEV therapy prevents antimicrobial resistance in nosocomial pathogens by eliminating off-target antibiotics. It is envisioned that SEV may protect DAP and VAN from resistance development, potentially addressing the long-lasting antimicrobial resistance.
Collapse
Affiliation(s)
- Roya Koshani
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Shang-Lin Yeh
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Zeming He
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Naveen Narasimhalu
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Landon G Vom Steeg
- Department of Biology and Entomology, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, 03756, USA
| | - Derek G Sim
- Department of Biology and Entomology, The Pennsylvania State University, University Park, PA, 16802, USA
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Robert J Woods
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Andrew F Read
- Department of Biology and Entomology, The Pennsylvania State University, University Park, PA, 16802, USA
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Amir Sheikhi
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Chemistry, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Neurosurgery, College of Medicine, The Pennsylvania State University, Hershey, PA, 17033, USA
| |
Collapse
|
23
|
Yilmaz B, Macpherson AJ. Delving the depths of 'terra incognita' in the human intestine - the small intestinal microbiota. Nat Rev Gastroenterol Hepatol 2025; 22:71-81. [PMID: 39443711 DOI: 10.1038/s41575-024-01000-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/25/2024] [Indexed: 10/25/2024]
Abstract
The small intestinal microbiota has a crucial role in gastrointestinal health, affecting digestion, immune function, bile acid homeostasis and nutrient metabolism. The challenges of accessibility at this site mean that our knowledge of the small intestinal microbiota is less developed than of the colonic or faecal microbiota. Here, we summarize the features and fluctuations of the microbiota along the small intestinal tract, focusing on humans, and discuss physicochemical factors and assessment methods, including the technical challenges of investigating the low microbial biomass of the proximal small bowel. We highlight the essential protective mechanisms of the small intestine, including motility, the paracellular barrier and mucus, and secretory immunity, to show their roles in limiting excessive exposure of host tissues to microbial metabolites. We address current knowledge gaps, particularly the variability among individuals, the effects of dysbiosis of the small intestinal microbiota on health and how different taxa in small intestinal microbiota could compensate for each other functionally.
Collapse
Affiliation(s)
- Bahtiyar Yilmaz
- Department of Visceral Surgery and Medicine, Bern University Hospital, University of Bern, Bern, Switzerland.
- Maurice Müller Laboratories, Department for Biomedical Research, University of Bern, Bern, Switzerland.
- Bern Center for Precision Medicine (BCPM), University of Bern, Bern, Switzerland.
| | - Andrew J Macpherson
- Department of Visceral Surgery and Medicine, Bern University Hospital, University of Bern, Bern, Switzerland.
- Maurice Müller Laboratories, Department for Biomedical Research, University of Bern, Bern, Switzerland.
- Bern Center for Precision Medicine (BCPM), University of Bern, Bern, Switzerland.
| |
Collapse
|
24
|
Lemmetty J, Lee Y, Laitila T, Bredehorst S, Coda R, Katina K, Maina NH. Sequestration of aflatoxin B1 by lactic acid bacteria: Role of binding and biotransformation. Food Res Int 2025; 199:115351. [PMID: 39658155 DOI: 10.1016/j.foodres.2024.115351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 12/12/2024]
Abstract
Climate change has introduced new challenges to food safety by altering the occurrence and distribution of fungi leading to increased mycotoxin contamination in crops. Among these mycotoxins, aflatoxin B1 (AFB1) stands out as a potent carcinogen, posing significant health risks to consumers. Methods for AFB1 decontamination have been intensively investigated and lactic acid bacteria (LAB) have gained increasing interest for their potential in AFB1 detoxification. The present study aimed to evaluate the potential of various LAB strains for reducing AFB1. A simple binding assay was used to evaluate the AFB1 binding by LAB. The ability of LAB to biotransform AFB1 was studied by simultaneous incubation of LAB and AFB1 in growth media and the biotransformation products were evaluated using liquid chromatography quadrupole time-of-flight mass spectrometry. Furthermore, the main reduction mechanism was investigated by conducting the binding assay using the same cell concentration used in the biotransformation experiment. The binding assay demonstrated a substantial reduction of AFB1 (16-71 %) depending on the strain, cell viability and pH. The binding capacity was shown to be strain-specific and improved when using non-viable cells. On the other hand, biotransformation exhibited much lower reduction of AFB1 (0-18 %). The reduction was also strain-specific and increased with longer incubation time. Additionally, no biotransformation products were found. The experiment on AFB1 reduction mechanism revealed binding as the primary mechanism of LAB for reducing AFB1, surpassing potential enzymatic degradation routes.
Collapse
Affiliation(s)
- Jenna Lemmetty
- Department of Food and Nutrition, University of Helsinki, Anges Sjöbergin katu 2, Helsinki 00790, Finland
| | - Youngsun Lee
- Department of Food and Nutrition, University of Helsinki, Anges Sjöbergin katu 2, Helsinki 00790, Finland.
| | - Tiina Laitila
- Department of Food and Nutrition, University of Helsinki, Anges Sjöbergin katu 2, Helsinki 00790, Finland
| | - Swantje Bredehorst
- Department of Food and Nutrition, University of Helsinki, Anges Sjöbergin katu 2, Helsinki 00790, Finland
| | - Rossana Coda
- Department of Food and Nutrition, University of Helsinki, Anges Sjöbergin katu 2, Helsinki 00790, Finland
| | - Kati Katina
- Department of Food and Nutrition, University of Helsinki, Anges Sjöbergin katu 2, Helsinki 00790, Finland
| | - Ndegwa H Maina
- Department of Food and Nutrition, University of Helsinki, Anges Sjöbergin katu 2, Helsinki 00790, Finland
| |
Collapse
|
25
|
Pouille CL, Dugardin C, Behra J, Tourret M, Molinié R, Fontaine JX, Mathiron D, Palaric C, Gagneul D, Ravallec R, Rambaud C, Hilbert JL, Lucau-Danila A, Cudennec B. Metabolomic monitoring of chicory during in vitro gastrointestinal digestion and correlation with bioactive properties. Food Chem 2024; 467:142344. [PMID: 39644662 DOI: 10.1016/j.foodchem.2024.142344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 11/21/2024] [Accepted: 12/01/2024] [Indexed: 12/09/2024]
Abstract
Chicory, recognized as a functional food, is increasingly becoming the focus of research. This study aimed to investigate the in vitro impact of gastrointestinal digestion on the composition and bioactive properties of chicory decoction. Chicory flour, derived from the roots, was transformed into an aqueous decoction and was subjected to simulated in vitro human gastrointestinal digestion (SGID). For the first time, the influence of the digestive process on specific classes of bioactive molecules was tracked across different digestive compartments (oral, gastric, and intestinal) using a metabolomic approach. Concurrently, the antioxidant, anti-inflammatory, and intestinal hormone regulation effects were assessed before and after SGID. The findings revealed that specific transformations of chlorogenic acid (CGA) and sesquiterpene lactones (STL) during SGID enhanced antioxidant and anti-inflammatory properties post-digestion. Quantitative results demonstrated a significant increase in ROS scavenging capacity and metabolite activity.
Collapse
Affiliation(s)
- Céline L Pouille
- Univ. Lille, UMRT 1158 BioEcoAgro, F-59000 Lille, France; Joint Laboratory CHIC41H University of Lille-Florimond-Desprez, Cité scientifique, 59655 Villeneuve d'Ascq, France
| | | | - Josette Behra
- Univ. Lille, UMRT 1158 BioEcoAgro, F-59000 Lille, France
| | | | - Roland Molinié
- UPJV, UMRT 1158 BioEcoAgro. BIOlogie des Plantes et Innovation (BIOPI), 80025 Amiens, France
| | - Jean-Xavier Fontaine
- UPJV, UMRT 1158 BioEcoAgro. BIOlogie des Plantes et Innovation (BIOPI), 80025 Amiens, France
| | - David Mathiron
- Plateforme Analytique UFR des Sciences, UPJV, Bâtiment Serres-Transfert Rue Dallery-Passage du Sourire d'Avril, 80039 Amiens, France
| | - Cécile Palaric
- Univ. Lille, UMRT 1158 BioEcoAgro, F-59000 Lille, France; Joint Laboratory CHIC41H University of Lille-Florimond-Desprez, Cité scientifique, 59655 Villeneuve d'Ascq, France
| | - David Gagneul
- Univ. Lille, UMRT 1158 BioEcoAgro, F-59000 Lille, France; Joint Laboratory CHIC41H University of Lille-Florimond-Desprez, Cité scientifique, 59655 Villeneuve d'Ascq, France
| | | | - Caroline Rambaud
- Univ. Lille, UMRT 1158 BioEcoAgro, F-59000 Lille, France; Joint Laboratory CHIC41H University of Lille-Florimond-Desprez, Cité scientifique, 59655 Villeneuve d'Ascq, France
| | - Jean-Louis Hilbert
- Univ. Lille, UMRT 1158 BioEcoAgro, F-59000 Lille, France; Joint Laboratory CHIC41H University of Lille-Florimond-Desprez, Cité scientifique, 59655 Villeneuve d'Ascq, France
| | - Anca Lucau-Danila
- Univ. Lille, UMRT 1158 BioEcoAgro, F-59000 Lille, France; Joint Laboratory CHIC41H University of Lille-Florimond-Desprez, Cité scientifique, 59655 Villeneuve d'Ascq, France.
| | | |
Collapse
|
26
|
Yu S, Webber MJ. Engineering disease analyte response in peptide self-assembly. J Mater Chem B 2024; 12:10757-10769. [PMID: 39382032 DOI: 10.1039/d4tb01860e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
A need to enhance the precision and specificity of therapeutic nanocarriers inspires the development of advanced nanomaterials capable of sensing and responding to disease-related cues. Self-assembled peptides offer a promising nanocarrier platform with versatile use to create precisely defined nanoscale materials. Disease-relevant cues can range from large biomolecules, such as enzymes, to ubiquitous small molecules with varying concentrations in healthy versus diseased states. Notably, pH changes (i.e., H+ concentration), redox species (e.g., H2O2), and glucose levels are significant spatial and/or temporal indicators of therapeutic need. Self-assembled peptides respond to these cues by altering their solubility, modulating electrostatic interactions, or facilitating chemical transformations through dynamic or labile bonds. This review explores the design and construction of therapeutic nanocarriers using self-assembled peptides, focusing on how peptide sequence engineering along with the inclusion of non-peptidic components can link the assembly state of these nanocarriers to the presence of disease-relevant small molecules.
Collapse
Affiliation(s)
- Sihan Yu
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Matthew J Webber
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| |
Collapse
|
27
|
Tollemeto M, Ursulska S, Welzen PLW, Thamdrup LHE, Malakpour-Permlid A, Li Y, Soufi G, Patiño Padial T, Christensen JB, Hagner Nielsen L, van Hest J, Boisen A. Tailored Polymersomes for Enhanced Oral Drug Delivery: pH-Sensitive Systems for Intestinal Delivery of Immunosuppressants. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2403640. [PMID: 38963162 DOI: 10.1002/smll.202403640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/11/2024] [Indexed: 07/05/2024]
Abstract
Ensuring precise drug release at target sites is crucial for effective treatment. Here, pH-responsive nanoparticles for oral administration of mycophenolate mofetil, an alternative therapy for patients with inflammatory bowel disease unresponsive to conventional treatments is developed. However, its oral administration presents challenges due to its low solubility in the small intestine and high solubility and absorption in the stomach. Therefore, this aim is to design a drug delivery system capable of maintaining drug solubility compared to the free drug while delaying absorption from the stomach to the intestine. Successful synthesis and assembly of a block copolymer incorporating a pH-responsive functional group is achieved. Dynamic light scattering indicated a significant change in hydrodynamic size when the pH exceeded 6.5, confirming successful incorporation of the pH-responsive group. Encapsulation and controlled release of mycophenolate mofetil are efficiently demonstrated, with 90% release observed at intestinal pH. In vitro cell culture studies confirmed biocompatibility, showing no toxicity or adverse effects on Caco-2 cells. In vivo oral rat studies indicated reduced drug absorption in the stomach and enhanced absorption in the small intestine with the developed formulation. This research presents a promising drug delivery system with potential applications in the treatment of inflammatory bowel disease.
Collapse
Affiliation(s)
- Matteo Tollemeto
- The Danish National Research Foundation and Villum Foundation's Center IDUN, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, 2800, Denmark
| | - Sintija Ursulska
- The Danish National Research Foundation and Villum Foundation's Center IDUN, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, 2800, Denmark
| | - Pascal L W Welzen
- Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, 5600 MB, The Netherlands
| | - Lasse H E Thamdrup
- The Danish National Research Foundation and Villum Foundation's Center IDUN, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, 2800, Denmark
| | - Atena Malakpour-Permlid
- The Danish National Research Foundation and Villum Foundation's Center IDUN, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, 2800, Denmark
| | - Yudong Li
- Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, 5600 MB, The Netherlands
| | - Gohar Soufi
- The Danish National Research Foundation and Villum Foundation's Center IDUN, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, 2800, Denmark
| | - Tania Patiño Padial
- Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, 5600 MB, The Netherlands
| | - Jørn B Christensen
- Department of Chemistry, University of Copenhagen, Thovaldsensvej 40, Frederiksberg, DK-1871, Denmark
| | - Line Hagner Nielsen
- The Danish National Research Foundation and Villum Foundation's Center IDUN, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, 2800, Denmark
| | - Jan van Hest
- Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, 5600 MB, The Netherlands
| | - Anja Boisen
- The Danish National Research Foundation and Villum Foundation's Center IDUN, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, 2800, Denmark
| |
Collapse
|
28
|
Karbasi AB, Barfuss JD, Morgan TC, Collins D, Costenbader DA, Dennis DG, Hinman A, Ko K, Messina C, Nguyen KC, Schugar RC, Stein KA, Williams BB, Xu H, Annes JP, Smith M. Sol-moiety: Discovery of a water-soluble prodrug technology for enhanced oral bioavailability of insoluble therapeutics. Nat Commun 2024; 15:8487. [PMID: 39353935 PMCID: PMC11445516 DOI: 10.1038/s41467-024-52793-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 09/23/2024] [Indexed: 10/03/2024] Open
Abstract
Though conceptually attractive, the use of water-soluble prodrug technology to enhance oral bioavailability of highly insoluble small molecule therapeutics has not been widely adopted. In large part, this is due to the rapid enzymatic or chemical hydrolysis of prodrugs within the gastrointestinal tract, resulting in drug precipitation and no overall improvement in oral bioavailability relative to standard formulation strategies. We reasoned that an optimal water-soluble prodrug could be attained if the rate of prodrug hydrolysis were reduced to favor drug absorption rather than drug precipitation. In doing so, the rate of hydrolysis provides a pharmacokinetic control point for drug delivery. Herein, we report the discovery of a water-soluble promoiety (Sol-moiety) technology to optimize the oral bioavailability of highly insoluble small molecule therapeutics, possessing various functional groups, without the need for sophisticated, often toxic, lipid or organic solvent-based formulations. The power of the technology is demonstrated with marked pharmacokinetic improvement of the commercial drugs enzalutamide, vemurafenib, and paclitaxel. This led to a successful efficacy study of a water-soluble orally administered prodrug of paclitaxel in a mouse pancreatic tumor model.
Collapse
Affiliation(s)
- Arvin B Karbasi
- Sarafan ChEM-H, Stanford University, Stanford, CA, 94305, USA
| | - Jaden D Barfuss
- Sarafan ChEM-H, Stanford University, Stanford, CA, 94305, USA
| | | | - Daniel Collins
- Sarafan ChEM-H, Stanford University, Stanford, CA, 94305, USA
| | | | - David G Dennis
- Sarafan ChEM-H, Stanford University, Stanford, CA, 94305, USA
| | - Andrew Hinman
- Sarafan ChEM-H, Stanford University, Stanford, CA, 94305, USA
| | - KyuWeon Ko
- Sarafan ChEM-H, Stanford University, Stanford, CA, 94305, USA
| | - Cynthia Messina
- Sarafan ChEM-H, Stanford University, Stanford, CA, 94305, USA
- Division of Endocrinology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Khanh C Nguyen
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Rebecca C Schugar
- Division of Endocrinology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Karin A Stein
- Sarafan ChEM-H, Stanford University, Stanford, CA, 94305, USA
| | | | - Haixia Xu
- Division of Endocrinology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Justin P Annes
- Division of Endocrinology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Mark Smith
- Sarafan ChEM-H, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
29
|
Malat I, Drancourt M, Grine G. Methanobrevibacter smithii cell variants in human physiology and pathology: A review. Heliyon 2024; 10:e36742. [PMID: 39347381 PMCID: PMC11437934 DOI: 10.1016/j.heliyon.2024.e36742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 08/14/2024] [Accepted: 08/21/2024] [Indexed: 10/01/2024] Open
Abstract
Methanobrevibacter smithii (M. smithii), initially isolated from human feces, has been recognised as a distinct taxon within the Archaea domain following comprehensive phenotypic, genetic, and genomic analyses confirming its uniqueness among methanogens. Its diversity, encompassing 15 genotypes, mirrors that of biotic and host-associated ecosystems in which M. smithii plays a crucial role in detoxifying hydrogen from bacterial fermentations, converting it into mechanically expelled gaseous methane. In microbiota in contact with host epithelial mucosae, M. smithii centres metabolism-driven microbial networks with Bacteroides, Prevotella, Ruminococcus, Veillonella, Enterococcus, Escherichia, Enterobacter, Klebsiella, whereas symbiotic association with the nanoarchaea Candidatus Nanopusillus phoceensis determines small and large cell variants of M. smithii. The former translocate with bacteria to induce detectable inflammatory and serological responses and are co-cultured from blood, urine, and tissular abscesses with bacteria, prototyping M. smithii as a model organism for pathogenicity by association. The sources, mechanisms and dynamics of in utero and lifespan M. smithii acquisition, its diversity, and its susceptibility to molecules of environmental, veterinary, and medical interest still have to be deeply investigated, as only four strains of M. smithii are available in microbial collections, despite the pivotal role this neglected microorganism plays in microbiota physiology and pathologies.
Collapse
Affiliation(s)
- Ihab Malat
- IHU Méditerranée Infection, Marseille, France
- Aix-Marseille-Université, MEPHI, IHU Méditerranée Infection, France
| | - Michel Drancourt
- IHU Méditerranée Infection, Marseille, France
- Aix-Marseille-Université, MEPHI, IHU Méditerranée Infection, France
| | - Ghiles Grine
- IHU Méditerranée Infection, Marseille, France
- Aix-Marseille-Université, MEPHI, IHU Méditerranée Infection, France
| |
Collapse
|
30
|
Ling J, Hryckowian AJ. Re-framing the importance of Group B Streptococcus as a gut-resident pathobiont. Infect Immun 2024; 92:e0047823. [PMID: 38436256 PMCID: PMC11392526 DOI: 10.1128/iai.00478-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024] Open
Abstract
Streptococcus agalactiae (Group B Streptococcus, GBS) is a Gram-positive bacterial species that causes disease in humans across the lifespan. While antibiotics are used to mitigate GBS infections, it is evident that antibiotics disrupt human microbiomes (which can predispose people to other diseases later in life), and antibiotic resistance in GBS is on the rise. Taken together, these unintended negative impacts of antibiotics highlight the need for precision approaches for minimizing GBS disease. One possible approach involves selectively depleting GBS in its commensal niches before it can cause disease at other body sites or be transmitted to at-risk individuals. One understudied commensal niche of GBS is the adult gastrointestinal (GI) tract, which may predispose colonization at other body sites in individuals at risk for GBS disease. However, a better understanding of the host-, microbiome-, and GBS-determined variables that dictate GBS GI carriage is needed before precise GI decolonization approaches can be developed. In this review, we synthesize current knowledge of the diverse body sites occupied by GBS as a pathogen and as a commensal. We summarize key molecular factors GBS utilizes to colonize different host-associated niches to inform future efforts to study GBS in the GI tract. We also discuss other GI commensals that are pathogenic in other body sites to emphasize the broader utility of precise de-colonization approaches for mitigating infections by GBS and other bacterial pathogens. Finally, we highlight how GBS treatments could be improved with a more holistic understanding of GBS enabled by continued GI-focused study.
Collapse
Affiliation(s)
- Joie Ling
- Department of
Medicine, Division of Gastroenterology and Hepatology, University of
Wisconsin School of Medicine and Public
Health, Madison,
Wisconsin, USA
- Department of Medical
Microbiology and Immunology, University of Wisconsin School of Medicine
and Public Healthon,
Madison, Wisconsin, USA
- Microbiology Doctoral
Training Program, University of
Wisconsin-Madison, Madison,
Wisconsin, USA
| | - Andrew J. Hryckowian
- Department of
Medicine, Division of Gastroenterology and Hepatology, University of
Wisconsin School of Medicine and Public
Health, Madison,
Wisconsin, USA
- Department of Medical
Microbiology and Immunology, University of Wisconsin School of Medicine
and Public Healthon,
Madison, Wisconsin, USA
| |
Collapse
|
31
|
Steigert S, Brouwers J, Verbeke K, Vanuytsel T, Augustijns P. Characterization of luminal contents from the fasted human proximal colon. Eur J Pharm Sci 2024; 200:106821. [PMID: 38823599 DOI: 10.1016/j.ejps.2024.106821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/03/2024]
Abstract
To treat colonic diseases more effectively, improved therapies are urgently needed. In this respect, delivering drugs locally to the colon is a key strategy to achieve higher local drug concentrations while minimizing systemic side effects. Understanding the luminal environment is crucial to efficiently develop such targeted therapies and to predict drug disposition in the colon. In this clinical study, we collected colonic contents from an undisturbed fasted proximal colon via colonoscopy and characterized their composition with regard to drug disposition. Colonic pH, osmolality, protein content, bile salts, lipids, phospholipids and short-chain fatty acids were investigated in 10 healthy volunteers (8 male and 2 female, age 19-25). The unique environment of the proximal colon was reflected in the composition of the sampled luminal fluids and the effect of the microbiota could be observed on the pH (median 6.55), the composition of bile salts (majority deconjugated and secondary), and the abundance of short-chain fatty acids. At the same time, an increase in phospholipid concentration, osmolality and total protein content compared to reported ileal values was seen, likely resulting from desiccation. Lipids could only be found in low quantities and mainly in the form of cholesterol and free fatty acids, showing almost complete digestion and absorption by the time luminal contents reach the colon. All characteristics also displayed the considerable intersubject variability found in different regions of the gastrointestinal tract. This study contributes to an improved understanding of the luminal conditions in the proximal colon and facilitates the development of new predictive tools to study colonic drug absorption.
Collapse
Affiliation(s)
- Sebastian Steigert
- Drug Delivery and Disposition, KU Leuven, Gasthuisberg O&N II, Herestraat 49 - box 921, 3000 Leuven, Belgium
| | - Joachim Brouwers
- Drug Delivery and Disposition, KU Leuven, Gasthuisberg O&N II, Herestraat 49 - box 921, 3000 Leuven, Belgium
| | - Kristin Verbeke
- Translational Research Center for Gastrointestinal Disorders, TARGID, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Tim Vanuytsel
- Translational Research Center for Gastrointestinal Disorders, TARGID, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Gastroenterology and Hepatology, University Hospitals Leuven campus Gasthuisberg, Herestraat 49, 3000 Leuven, Belgium
| | - Patrick Augustijns
- Drug Delivery and Disposition, KU Leuven, Gasthuisberg O&N II, Herestraat 49 - box 921, 3000 Leuven, Belgium.
| |
Collapse
|
32
|
Guo Y, Ren C, He Y, Wu Y, Yang X. Deciphering the spatiotemporal transcriptional landscape of intestinal diseases (Review). Mol Med Rep 2024; 30:157. [PMID: 38994768 PMCID: PMC11258600 DOI: 10.3892/mmr.2024.13281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 04/19/2024] [Indexed: 07/13/2024] Open
Abstract
The intestines are the largest barrier organ in the human body. The intestinal barrier plays a crucial role in maintaining the balance of the intestinal environment and protecting the intestines from harmful bacterial invasion. Single‑cell RNA sequencing technology allows the detection of the different cell types in the intestine in two dimensions and the exploration of cell types that have not been fully characterized. The intestinal mucosa is highly complex in structure, and its proper functioning is linked to multiple structures in the proximal‑distal intestinal and luminal‑mucosal axes. Spatial localization is at the core of the efforts to explore the interactions between the complex structures. Spatial transcriptomics (ST) is a method that allows for comprehensive tissue analysis and the acquisition of spatially separated genetic information from individual cells, while preserving their spatial location and interactions. This approach also prevents the loss of fragile cells during tissue disaggregation. The emergence of ST technology allows us to spatially dissect enzymatic processes and interactions between multiple cells, genes, proteins and signals in the intestine. This includes the exchange of oxygen and nutrients in the intestine, different gradients of microbial populations and the role of extracellular matrix proteins. This regionally precise approach to tissue studies is gaining more acceptance and is increasingly applied in the investigation of disease mechanisms related to the gastrointestinal tract. Therefore, this review summarized the application of ST in gastrointestinal diseases.
Collapse
Affiliation(s)
- Yajing Guo
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, P.R. China
| | - Chao Ren
- Graduate School, Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Yuxi He
- Department of Digestive Medicine, Chongqing City Hospital of Traditional Chinese Medicine, Chongqing 400021, P.R. China
| | - Yue Wu
- Department of Digestive Medicine, Chongqing City Hospital of Traditional Chinese Medicine, Chongqing 400021, P.R. China
| | - Xiaojun Yang
- Department of Digestive Medicine, Chongqing City Hospital of Traditional Chinese Medicine, Chongqing 400021, P.R. China
| |
Collapse
|
33
|
Li Z, Huang Z, Gu P. Response of Escherichia coli to Acid Stress: Mechanisms and Applications-A Narrative Review. Microorganisms 2024; 12:1774. [PMID: 39338449 PMCID: PMC11434309 DOI: 10.3390/microorganisms12091774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
Change in pH in growth conditions is the primary stress for most neutralophilic bacteria, including model microorganism Escherichia coli. However, different survival capacities under acid stress in different bacteria are ubiquitous. Research on different acid-tolerance mechanisms in microorganisms is important for the field of combating harmful gut bacteria and promoting fermentation performance of industrial strains. Therefore, this study aimed to carry out a narrative review of acid-stress response mechanism of E. coli discovered so far, including six AR systems, cell membrane protection, and macromolecular repair. In addition, the application of acid-tolerant E. coli in industry was illustrated, such as production of industrial organic acid and developing bioprocessing for industrial wastes. Identifying these aspects will open the opportunity for discussing development aspects for subsequent research of acid-tolerant mechanisms and application in E. coli.
Collapse
Affiliation(s)
| | | | - Pengfei Gu
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China; (Z.L.); (Z.H.)
| |
Collapse
|
34
|
Verma P, Rezaei L, Govindarajan R, Greig NH, Donovan MD. Gastroretentive Delivery Approach to Address pH-Dependent Degradation of (+)- and (-)-Phenserine. AAPS PharmSciTech 2024; 25:198. [PMID: 39192157 DOI: 10.1208/s12249-024-02903-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 07/26/2024] [Indexed: 08/29/2024] Open
Abstract
(-)-Phenserine ("phenserine") and (+)-phenserine (posiphen; buntanetap) are longer-acting enantiomeric analogs of physostigmine with demonstrated promise in the treatment of Alzheimer's and Parkinson's diseases. Both enantiomers have short plasma half-lives, and their pharmacokinetics might be improved through the use of either once or twice-daily administration of an extended-release dosage form. Phenserine was observed to form a colored degradation product in near-neutral and alkaline pH environments, and at pH 7, the half-life of posiphen was determined to be ~ 9 h (40 °C). To limit luminal degradation which would reduce bioavailability, a gastroretentive tablet composed of a polyethylene oxide-xanthan gum matrix was developed. When placed in simulated gastric fluid (pH 1.2), approximately 70% of the phenserine was released over a 12 h period, and no degradants were detected in the release medium. In comparison, a traditional hydrophilic-matrix, extended-release tablet showed measurable amounts of phenserine degradation in a pH 7.2 medium over an 8 h release interval. These results confirm that a gastroretentive tablet can reduce the luminal degradation of phenserine or posiphen by limiting exposure to neutral pH conditions while providing sustained release of the drug over at least 12 h. Additional advantages of the gastroretentive tablet include reduced gastric and intestinal concentrations of the drug resulting from the slower release from the gastroretentive tablet which may also limit the occurrence of the dose-limiting GI side effects previously observed with immediate-release phenserine capsules.
Collapse
Affiliation(s)
- Pratishtha Verma
- Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Leyla Rezaei
- Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Ramprakash Govindarajan
- Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Nigel H Greig
- Translational Gerontology Branch, National Institute On Aging, Intramural Research Program, National Institutes of Health, Baltimore, Maryland, 21224, USA
| | - Maureen D Donovan
- Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa, Iowa City, Iowa, 52242, USA.
| |
Collapse
|
35
|
Gulkis M, Luo M, Chipman P, Mietzsch M, Söderlund-Venermo M, Bennett A, McKenna R. Structural Characterization of Human Bufavirus 1: Receptor Binding and Endosomal pH-Induced Changes. Viruses 2024; 16:1258. [PMID: 39205232 PMCID: PMC11360561 DOI: 10.3390/v16081258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/30/2024] [Accepted: 08/03/2024] [Indexed: 09/04/2024] Open
Abstract
Bufaviruses (BuV) are members of the Parvoviridae of the Protoparvovirus genus. They are non-enveloped, T = 1 icosahedral ssDNA viruses isolated from patients exhibiting acute diarrhea. The lack of treatment options and a limited understanding of their disease mechanisms require studying these viruses on a molecular and structural level. In the present study, we utilize glycan arrays and cell binding assays to demonstrate that BuV1 capsid binds terminal sialic acid (SIA) glycans. Furthermore, using cryo-electron microscopy (cryo-EM), SIA is shown to bind on the 2/5-fold wall of the capsid surface. Interestingly, the capsid residues stabilizing SIA binding are conserved in all human BuVs identified to date. Additionally, biophysical assays illustrate BuV1 capsid stabilization during endo-lysosomal (pH 7.4-pH 4) trafficking and capsid destabilization at pH 3 and less, which correspond to the pH of the stomach. Hence, we determined the cryo-EM structures of BuV1 capsids at pH 7.4, 4.0, and 2.6 to 2.8 Å, 3.2 Å, and 2.7 Å, respectively. These structures reveal capsid structural rearrangements during endo-lysosomal escape and provide a potential mechanism for this process. The structural insights gained from this study will add to the general knowledge of human pathogenic parvoviruses. Furthermore, the identification of the conserved SIA receptor binding site among BuVs provides a possible targetable surface-accessible pocket for the design of small molecules to be developed as anti-virals for these viruses.
Collapse
Affiliation(s)
- Mitchell Gulkis
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32611, USA; (M.G.); (M.L.); (P.C.); (M.M.)
| | - Mengxiao Luo
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32611, USA; (M.G.); (M.L.); (P.C.); (M.M.)
| | - Paul Chipman
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32611, USA; (M.G.); (M.L.); (P.C.); (M.M.)
| | - Mario Mietzsch
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32611, USA; (M.G.); (M.L.); (P.C.); (M.M.)
| | - Maria Söderlund-Venermo
- Department of Virology, University of Helsinki, P.O. Box 21 (Haartmaninkatu 3), FIN-00014 Helsinki, Finland;
| | - Antonette Bennett
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32611, USA; (M.G.); (M.L.); (P.C.); (M.M.)
| | - Robert McKenna
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32611, USA; (M.G.); (M.L.); (P.C.); (M.M.)
| |
Collapse
|
36
|
Abbasi A, Bazzaz S, Da Cruz AG, Khorshidian N, Saadat YR, Sabahi S, Ozma MA, Lahouty M, Aslani R, Mortazavian AM. A Critical Review on Akkermansia muciniphila: Functional Mechanisms, Technological Challenges, and Safety Issues. Probiotics Antimicrob Proteins 2024; 16:1376-1398. [PMID: 37432597 DOI: 10.1007/s12602-023-10118-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2023] [Indexed: 07/12/2023]
Abstract
Due to its physiological benefits from in vitro and in vivo points of view, Akkermansia muciniphila, a common colonizer in the human gut mucous layer, has consistently been identified as an option for the next-generation probiotic. A. muciniphila is a significant bacterium that promotes host physiology. However, it also has a great deal of potential to become a probiotic due to its physiological advantages in a variety of therapeutic circumstances. Therefore, it can be established that the abundance of A. muciniphila in the gut environment, which is controlled by many genetic and dietary variables, is related to the biological behaviors of the intestinal microbiota and gut dysbiosis/eubiosis circumstances. Before A. muciniphila is widely utilized as a next-generation probiotic, regulatory obstacles, the necessity for significant clinical trials, and the sustainability of manufacturing must be eliminated. In this review, the outcomes of recent experimental and clinical reports are comprehensively reviewed, and common colonization patterns, main factors involved in the colonization of A. muciniphila in the gut milieu, their functional mechanisms in establishing homeostasis in the metabolic and energy pathways, the promising delivery role of microencapsulation, potential genetic engineering strategies, and eventually safety issues of A. muciniphila have been discussed.
Collapse
Affiliation(s)
- Amin Abbasi
- Student Research Committee, Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Bazzaz
- Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Adriano G Da Cruz
- Department of Food Processing, Federal Institute of Science and Technology Education of Rio de Janeiro (IFRJ) - Campus Maracanã, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Nasim Khorshidian
- Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Sahar Sabahi
- Department of Nutrition, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mahdi Asghari Ozma
- Department of Medical Bacteriology and Virology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoud Lahouty
- Department of Microbiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Ramin Aslani
- Food Safety and Hygiene Division, Department of Environmental Health Engineering, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir M Mortazavian
- Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
37
|
Woodward SE, Neufeld LMP, Peña-Díaz J, Feng W, Serapio-Palacios A, Tarrant I, Deng W, Finlay BB. Both pathogen and host dynamically adapt pH responses along the intestinal tract during enteric bacterial infection. PLoS Biol 2024; 22:e3002761. [PMID: 39146372 PMCID: PMC11349234 DOI: 10.1371/journal.pbio.3002761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 08/27/2024] [Accepted: 07/19/2024] [Indexed: 08/17/2024] Open
Abstract
Enteric pathogens navigate distinct regional microenvironments within the intestine that cue important adaptive behaviors. We investigated the response of Citrobacter rodentium, a model of human pathogenic Escherichia coli infection in mice, to regional gastrointestinal pH. We found that small intestinal pH (4.4-4.8) triggered virulence gene expression and altered cell morphology, supporting initial intestinal attachment, while higher pH, representative of C. rodentium's replicative niches further along the murine intestine, supported pathogen growth. Gastric pH, a key barrier to intestinal colonization, caused significant accumulation of intra-bacterial reactive oxygen species (ROS), inhibiting growth of C. rodentium and related human pathogens. Within-host adaptation increased gastric acid survival, which may be due to a robust acid tolerance response (ATR) induced at colonic pH. However, the intestinal environment changes throughout the course of infection. We found that murine gastric pH decreases postinfection, corresponding to increased serum gastrin levels and altered host expression of acid secretion-related genes. Similar responses following Salmonella infection may indicate a protective host response to limit further pathogen ingestion. Together, we highlight interlinked bacterial and host adaptive pH responses as an important component of host-pathogen coevolution.
Collapse
Affiliation(s)
- Sarah E. Woodward
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, Canada
| | - Laurel M. P. Neufeld
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
| | - Jorge Peña-Díaz
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, Canada
| | - Wenny Feng
- Michael Smith Laboratories, University of British Columbia, Vancouver, Canada
| | - Antonio Serapio-Palacios
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, Canada
| | - Isabel Tarrant
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, Canada
| | - Wanyin Deng
- Michael Smith Laboratories, University of British Columbia, Vancouver, Canada
| | - B. Brett Finlay
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| |
Collapse
|
38
|
Muramatsu MK, Winter SE. Nutrient acquisition strategies by gut microbes. Cell Host Microbe 2024; 32:863-874. [PMID: 38870902 PMCID: PMC11178278 DOI: 10.1016/j.chom.2024.05.011] [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: 03/25/2024] [Revised: 05/08/2024] [Accepted: 05/14/2024] [Indexed: 06/15/2024]
Abstract
The composition and function of the gut microbiota are intimately tied to nutrient acquisition strategies and metabolism, with significant implications for host health. Both dietary and host-intrinsic factors influence community structure and the basic modes of bacterial energy metabolism. The intestinal tract is rich in carbon and nitrogen sources; however, limited access to oxygen restricts energy-generating reactions to fermentation. By contrast, increased availability of electron acceptors during episodes of intestinal inflammation results in phylum-level changes in gut microbiota composition, suggesting that bacterial energy metabolism is a key driver of gut microbiota function. In this review article, we will illustrate diverse examples of microbial nutrient acquisition strategies in the context of habitat filters and anatomical location and the central role of energy metabolism in shaping metabolic strategies to support bacterial growth in the mammalian gut.
Collapse
Affiliation(s)
- Matthew K Muramatsu
- Department of Internal Medicine, Division of Infectious Diseases, UC Davis, Davis, CA 95616, USA
| | - Sebastian E Winter
- Department of Internal Medicine, Division of Infectious Diseases, UC Davis, Davis, CA 95616, USA.
| |
Collapse
|
39
|
Yamamoto H, Sugano K. Drug Crystal Precipitation in Biorelevant Bicarbonate Buffer: A Well-Controlled Comparative Study with Phosphate Buffer. Mol Pharm 2024; 21:2854-2864. [PMID: 38718215 DOI: 10.1021/acs.molpharmaceut.4c00028] [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] [Indexed: 06/04/2024]
Abstract
The purpose of the present study was to clarify whether the precipitation profile of a drug in bicarbonate buffer (BCB) may differ from that in phosphate buffer (PPB) by a well-controlled comparative study. The precipitation profiles of structurally diverse poorly soluble drugs in BCB and PPB were evaluated by a pH-shift precipitation test or a solvent-shift precipitation test (seven weak acid drugs (pKa: 4.2 to 7.5), six weak base drugs (pKa: 4.8 to 8.4), one unionizable drug, and one zwitterionic drug). To focus on crystal precipitation processes, each ionizable drug was first completely dissolved in an HCl (pH 3.0) or NaOH (pH 11.0) aqueous solution (450 mL, 50 rpm, 37 °C). A 10-fold concentrated buffer solution (50 mL) was then added to shift the pH value to 6.5 to initiate precipitation (final volume: 500 mL, buffer capacity (β): 4.4 mM/ΔpH (BCB: 10 mM or PPB: 8 mM), ionic strength (I): 0.14 M (adjusted by NaCl)). The pH, β, and I values were set to be relevant to the physiology of the small intestine. For an unionizable drug, a solvent-shift method was used (1/100 dilution). To maintain the pH value of BCB, a floating lid was used to avoid the loss of CO2. The floating lid was applied also to PPB to precisely align the experimental conditions between BCB and PPB. The solid form of the precipitants was identified by powder X-ray diffraction and differential scanning microscopy. The precipitation of weak acids (pKa ≤ 5.1) and weak bases (pKa ≥ 7.3) was found to be slower in BCB than in PPB. In contrast, the precipitation profiles in BCB and PPB were similar for less ionizable or nonionizable drugs at pH 6.5. The final pH values of the bulk phase were pH 6.5 ± 0.1 after the precipitation tests in all cases. All precipitates were in their respective free forms. The precipitation of ionizable weak acids and bases was slower in BCB than in PPB. The surface pH of precipitating particles may have differed between BCB and PPB due to the slow hydration process of CO2 specific to BCB. Since BCB is a physiological buffer in the small intestine, it should be considered as an option for precipitation studies of ionizable weak acids and bases.
Collapse
Affiliation(s)
- Hibiki Yamamoto
- Molecular Pharmaceutics Laboratory, College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1, Noji-higashi, Kusatsu, Shiga 525-8577, Japan
| | - Kiyohiko Sugano
- Molecular Pharmaceutics Laboratory, College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1, Noji-higashi, Kusatsu, Shiga 525-8577, Japan
| |
Collapse
|
40
|
Wan W, Wu W, Amier Y, Li X, Yang J, Huang Y, Xun Y, Yu X. Engineered microorganisms: A new direction in kidney stone prevention and treatment. Synth Syst Biotechnol 2024; 9:294-303. [PMID: 38510204 PMCID: PMC10950756 DOI: 10.1016/j.synbio.2024.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/31/2024] [Accepted: 02/20/2024] [Indexed: 03/22/2024] Open
Abstract
Numerous studies have shown that intestinal and urinary tract flora are closely related to the formation of kidney stones. The removal of probiotics represented by lactic acid bacteria and the colonization of pathogenic bacteria can directly or indirectly promote the occurrence of kidney stones. However, currently existing natural probiotics have limitations. Synthetic biology is an emerging discipline in which cells or living organisms are genetically designed and modified to have biological functions that meet human needs, or even create new biological systems, and has now become a research hotspot in various fields. Using synthetic biology approaches of microbial engineering and biological redesign to enable probiotic bacteria to acquire new phenotypes or heterologous protein expression capabilities is an important part of synthetic biology research. Synthetic biology modification of microorganisms in the gut and urinary tract can effectively inhibit the development of kidney stones by a range of means, including direct degradation of metabolites that promote stone production or indirect regulation of flora homeostasis. This article reviews the research status of engineered microorganisms in the prevention and treatment of kidney stones, to provide a new and effective idea for the prevention and treatment of kidney stones.
Collapse
Affiliation(s)
- Wenlong Wan
- Department of Urology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Weisong Wu
- Department of Urology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yirixiatijiang Amier
- Department of Urology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xianmiao Li
- Department of Urology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Junyi Yang
- Department of Urology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yisheng Huang
- Department of Urology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yang Xun
- Department of Urology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xiao Yu
- Department of Urology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
41
|
Adamberg S, Adamberg K. Prevotella enterotype associates with diets supporting acidic faecal pH and production of propionic acid by microbiota. Heliyon 2024; 10:e31134. [PMID: 38779015 PMCID: PMC11109898 DOI: 10.1016/j.heliyon.2024.e31134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 03/21/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
Metabolism of dietary fibres by colon microbiota plays an important role for human health. Personal data from a nutrition study (57 subjects) were analysed to elucidate quantitative associations between the diet, faecal microbiome, organic acid concentrations and pH. Ratios of the predominant acids acetate, butyrate and propionate ranged from 1:0.67:0.27 to 1:0.17:0.36. Pectin-rich diets resulted in higher faecal acetate concentrations. Negative correlation between faecal pH and BSS was observed. Higher faecal pH and lower acid concentrations were related to the higher abundance of amino acid degrading Clostridium, Odoribacter and Eubacterium coprostanoligenes, which are weak carbohydrate fermenting taxa. Propionic acid correlated especially to high abundance of Prevotella and low abundance of proteobacteria. The acetate to propionate ratio of the Prevotella enterotype was about half of that of the Bacteroides enterotype. Based on the results we suggest the measurement of faecal pH and organic acid composition for research and diagnostic purposes.
Collapse
Affiliation(s)
- Signe Adamberg
- Tallinn University of Technology, Department of Chemistry and Biotechnology, 12618, Tallinn, Estonia
| | - Kaarel Adamberg
- Tallinn University of Technology, Department of Chemistry and Biotechnology, 12618, Tallinn, Estonia
- Center of Food and Fermentation Technologies, 12618, Tallinn, Estonia
| |
Collapse
|
42
|
Kadian S, Gopalakrishnan S, Selvamani V, Khan S, Meyer T, Thomas R, Rana MM, Irazoqui PP, Verma MS, Rahimi R. Smart Capsule for Targeted Detection of Inflammation Levels Inside the GI Tract. IEEE Trans Biomed Eng 2024; 71:1565-1576. [PMID: 38096093 PMCID: PMC11187759 DOI: 10.1109/tbme.2023.3343337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Effective management of Inflammatory Bowel Disease (IBD) is contingent upon frequent monitoring of inflammation levels at targeted locations within the gastrointestinal (GI) tract. This is crucial for assessing disease progression and detecting potential relapses. To address this need, a novel single-use capsule technology has been devised that enables region-specific inflammation measurement, thereby facilitating repeatable monitoring within the GI tract. The capsule integrates a pH-responsive coating for location-specific activation, a chemiluminescent paper-based myeloperoxidase (MPO) sensor for inflammation detection, and a miniaturized photodetector, complemented by embedded electronics for real-time wireless data transmission. Demonstrating linear sensitivity within the physiological MPO concentration range, the sensor is capable of effectively identifying inflammation risk in the GI fluid. Luminescence emitted by the sensor, proportional to MPO concentration, is converted into an electrical signal by the photodetector, generating a quantifiable energy output with a sensitivity of 6.14 µJ/U.ml-1. The capsule was also tested with GI fluids collected from pig models simulating various inflammation states. Despite the physiological complexities, the capsule consistently activated in the intended region and accurately detected MPO levels with less than a 5% variation between readings in GI fluid and a PBS solution. This study heralds a significant step towards minimally invasive, in situ GI inflammation monitoring, potentially revolutionizing personalized IBD management and patient-specific therapeutic strategies.
Collapse
|
43
|
Seco A, Pereira AR, Camuenho A, Oliveira J, Dias R, Brás N, Basílio N, Parola AJ, Lima JC, de Freitas V, Pina F. Comparing the Chemistry of Malvidin-3- O-glucoside and Malvidin-3,5- O-diglucoside Networks: A Holistic Approach to the Acidic and Basic Paradigms with Implications in Biological Studies. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:7497-7510. [PMID: 38520401 PMCID: PMC10995998 DOI: 10.1021/acs.jafc.4c00552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/07/2024] [Accepted: 03/11/2024] [Indexed: 03/25/2024]
Abstract
The kinetics, thermodynamics, and degradation of malvidin mono- and diglucosides were studied following a holistic approach by extending to the basic medium. In acidic conditions, the reversible kinetics of the flavylium cation toward the equilibrium is controlled by the hydration and cis-trans isomerization steps, while in the basic medium, the OH- nucleophilic addition to the anionic quinoidal bases is the slowest step. There is a pH range (transition pHs), between the acidic and basic paradigms, that includes physiological pH (7.4), where degradation reactions occur faster, preventing the system from reaching the equilibrium. The transition pH of the diglucoside is narrower, and in contrast with the monoglucoside, there is no evidence for the formation of colored oligomers among the degradation products. Noteworthy, OH- addition in position 4 to form B42-, a kinetic product that decreases the overall equilibration rate, was observed only for the diglucoside.
Collapse
Affiliation(s)
- André Seco
- LAQV—REQUIMTE,
Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal
| | - Ana Rita Pereira
- LAQV—REQUIMTE,
Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre, 687, 4169-007 Porto, Portugal
| | - Ambrósio Camuenho
- LAQV—REQUIMTE,
Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal
| | - Joana Oliveira
- LAQV—REQUIMTE,
Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre, 687, 4169-007 Porto, Portugal
| | - Ricardo Dias
- LAQV—REQUIMTE,
Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre, 687, 4169-007 Porto, Portugal
| | - Natércia
F. Brás
- LAQV—REQUIMTE,
Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre, 687, 4169-007 Porto, Portugal
| | - Nuno Basílio
- LAQV—REQUIMTE,
Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal
| | - A. Jorge Parola
- LAQV—REQUIMTE,
Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal
| | - João C. Lima
- LAQV—REQUIMTE,
Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal
| | - Victor de Freitas
- LAQV—REQUIMTE,
Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre, 687, 4169-007 Porto, Portugal
| | - Fernando Pina
- LAQV—REQUIMTE,
Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal
| |
Collapse
|
44
|
Blaesi AH, Saka N. WITHDRAWN: Gastroretentive fibrous dosage forms for prolonged delivery of sparingly soluble tyrosine kinase inhibitors. Part 3: Theoretical models of in vivo expansion, gastric residence time, and drug concentration in blood. Int J Pharm 2024; 653:123478. [PMID: 37839493 DOI: 10.1016/j.ijpharm.2023.123478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 09/25/2023] [Accepted: 10/01/2023] [Indexed: 10/17/2023]
Affiliation(s)
- Aron H Blaesi
- Enzian Pharmaceutics Aron H. Blaesi, CH-7078, Lenzerheide, Switzerland; Enzian Pharmaceutics, Inc., Cambridge, MA, 02139, USA.
| | - Nannaji Saka
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| |
Collapse
|
45
|
Zhou Y, Yao Y, Zhai Z, Mohamed MA, Mazzini F, Qi Q, Bortner MJ, Taylor LS, Edgar KJ. Reductive amination of oxidized hydroxypropyl cellulose with ω-aminoalkanoic acids as an efficient route to zwitterionic derivatives. Carbohydr Polym 2024; 328:121699. [PMID: 38220336 DOI: 10.1016/j.carbpol.2023.121699] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 12/02/2023] [Accepted: 12/13/2023] [Indexed: 01/16/2024]
Abstract
Zwitterionic polymers, with their equal amounts of cationic and anionic functional groups, have found widespread utility including as non-fouling coatings, hydrogel materials, stabilizers, antifreeze materials, and drug carriers. Polysaccharide-derived zwitterionic polymers are attractive because of their sustainable origin, potential for lower toxicity, and possible biodegradability, but previous methods for synthesis of zwitterionic polysaccharide derivatives have been limited in terms of flexibility and attainable degree of substitution (DS) of charged entities. We report herein successful design and synthesis of zwitterionic polysaccharide derivatives, in this case based on cellulose, by reductive amination of oxidized 2-hydroxypropyl cellulose (Ox-HPC) with ω-aminoalkanoic acids. Reductive amination products could be readily obtained with DS(cation) (= DS(anion)) up to 1.6. Adduct hydrophilic/hydrophobic balance (amphiphilicity) can be influenced by selecting the appropriate chain length of the ω-aminoalkanoic acid. This strategy is shown to produce a range of amphiphilic, water-soluble, moderately high glass transition temperature (Tg) polysaccharide derivatives in just a couple of efficient steps from commercially available building blocks. The adducts were evaluated as crystallization inhibitors. They are strong inhibitors of crystallization even for the challenging, poorly soluble, fast-crystallizing prostate cancer drug enzalutamide, as supported by surface tension and Flory-Huggins interaction parameter results.
Collapse
Affiliation(s)
- Yang Zhou
- Department of Sustainable Biomaterials, Virginia Tech, Blacksburg, VA 24061, United States; Macromolecules Innovation Institute, Virginia Tech, Blacksburg, VA 24061, United States; Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, United States.
| | - Yimin Yao
- Department of Chemical Engineering, Virginia Tech, Blacksburg, VA 24061, United States; Macromolecules Innovation Institute, Virginia Tech, Blacksburg, VA 24061, United States
| | - Zhenghao Zhai
- Macromolecules Innovation Institute, Virginia Tech, Blacksburg, VA 24061, United States
| | - Mennatallah A Mohamed
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, IN 47907, United States
| | - Fiorella Mazzini
- Macromolecules Innovation Institute, Virginia Tech, Blacksburg, VA 24061, United States
| | - Qingqing Qi
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, IN 47907, United States
| | - Michael J Bortner
- Department of Chemical Engineering, Virginia Tech, Blacksburg, VA 24061, United States; Macromolecules Innovation Institute, Virginia Tech, Blacksburg, VA 24061, United States
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, IN 47907, United States
| | - Kevin J Edgar
- Department of Sustainable Biomaterials, Virginia Tech, Blacksburg, VA 24061, United States; Macromolecules Innovation Institute, Virginia Tech, Blacksburg, VA 24061, United States
| |
Collapse
|
46
|
Mehrotra S, Kalyan BG P, Nayak PG, Joseph A, Manikkath J. Recent Progress in the Oral Delivery of Therapeutic Peptides and Proteins: Overview of Pharmaceutical Strategies to Overcome Absorption Hurdles. Adv Pharm Bull 2024; 14:11-33. [PMID: 38585454 PMCID: PMC10997937 DOI: 10.34172/apb.2024.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 07/04/2023] [Accepted: 08/16/2023] [Indexed: 04/09/2024] Open
Abstract
Purpose Proteins and peptides have secured a place as excellent therapeutic moieties on account of their high selectivity and efficacy. However due to oral absorption limitations, current formulations are mostly delivered parenterally. Oral delivery of peptides and proteins (PPs) can be considered the need of the hour due to the immense benefits of this route. This review aims to critically examine and summarize the innovations and mechanisms involved in oral delivery of peptide and protein drugs. Methods Comprehensive literature search was undertaken, spanning the early development to the current state of the art, using online search tools (PubMed, Google Scholar, ScienceDirect and Scopus). Results Research in oral delivery of proteins and peptides has a rich history and the development of biologics has encouraged additional research effort in recent decades. Enzyme hydrolysis and inadequate permeation into intestinal mucosa are the major causes that result in limited oral absorption of biologics. Pharmaceutical and technological strategies including use of absorption enhancers, enzyme inhibition, chemical modification (PEGylation, pro-drug approach, peptidomimetics, glycosylation), particulate delivery (polymeric nanoparticles, liposomes, micelles, microspheres), site-specific delivery in the gastrointestinal tract (GIT), membrane transporters, novel approaches (self-nanoemulsifying drug delivery systems, Eligen technology, Peptelligence, self-assembling bubble carrier approach, luminal unfolding microneedle injector, microneedles) and lymphatic targeting, are discussed. Limitations of these strategies and possible innovations for improving oral bioavailability of protein and peptide drugs are discussed. Conclusion This review underlines the application of oral route for peptide and protein delivery, which can direct the formulation scientist for better exploitation of this route.
Collapse
Affiliation(s)
- Sonal Mehrotra
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Pavan Kalyan BG
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Pawan Ganesh Nayak
- Department of Pharmacology,Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | | | - Jyothsna Manikkath
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| |
Collapse
|
47
|
Amekyeh H, Sabra R, Billa N. A Window for Enhanced Oral Delivery of Therapeutics via Lipid Nanoparticles. Drug Des Devel Ther 2024; 18:613-630. [PMID: 38476206 PMCID: PMC10927375 DOI: 10.2147/dddt.s439975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 11/25/2023] [Indexed: 03/14/2024] Open
Abstract
Oral administration of dosage forms is convenient and beneficial in several respects. Lipid nanoparticulate dosage forms have emerged as a useful carrier system in deploying low solubility drugs systemically, particularly class II, III, and IV drugs of the Biopharmaceutics Classification System. Like other nanoparticulate delivery systems, their low size-to-volume ratio facilitates uptake by phagocytosis. Lipid nanoparticles also provide scope for high drug loading and extended-release capability, ensuring diminished systemic side effects and improved pharmacokinetics. However, rapid gastrointestinal (GI) clearance of particulate delivery systems impedes efficient uptake across the mucosa. Mucoadhesion of dosage forms to the GI mucosa results in longer transit times due to interactions between the former and mucus. Delayed transit times facilitate transfer of the dosage form across the mucosa. In this regard, a balance between mucoadhesion and mucopenetration guarantees optimal systemic transfer. Furthermore, the interplay between GI anatomy and physiology is key to ensuring efficient systemic uptake. This review captures salient anatomical and physiological features of the GI tract and how these can be exploited for maximal systemic delivery of lipid nanoparticles. Materials used to impart mucoadhesion and examples of successful mucoadhesive lipid nanoformulations are highlighted in this review.
Collapse
Affiliation(s)
- Hilda Amekyeh
- Department of Pharmaceutics, School of Pharmacy, University of Health and Allied Sciences, Ho, Ghana
| | - Rayan Sabra
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
| | | |
Collapse
|
48
|
Padilla L, Fricker AD, Luna E, Choudhury B, Hughes ER, Panzetta ME, Valdivia RH, Flores GE. Mechanism of 2'-fucosyllactose degradation by human-associated Akkermansia. J Bacteriol 2024; 206:e0033423. [PMID: 38299857 PMCID: PMC10886448 DOI: 10.1128/jb.00334-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 01/04/2024] [Indexed: 02/02/2024] Open
Abstract
Among the first microorganisms to colonize the human gut of breastfed infants are bacteria capable of fermenting human milk oligosaccharides (HMOs). One of the most abundant HMOs, 2'-fucosyllactose (2'-FL), may specifically drive bacterial colonization of the intestine. Recently, differential growth has been observed across multiple species of Akkermansia on various HMOs including 2'-FL. In culture, we found growth of two species, A. muciniphila MucT and A. biwaensis CSUN-19,on HMOs corresponded to a decrease in the levels of 2'-FL and an increase in lactose, indicating that the first step in 2'-FL catabolism is the cleavage of fucose. Using phylogenetic analysis and transcriptional profiling, we found that the number and expression of fucosidase genes from two glycoside hydrolase (GH) families, GH29 and GH95, vary between these two species. During the mid-log phase of growth, the expression of several GH29 genes was increased by 2'-FL in both species, whereas the GH95 genes were induced only in A. muciniphila. We further show that one putative fucosidase and a β-galactosidase from A. biwaensis are involved in the breakdown of 2'-FL. Our findings indicate that the plasticity of GHs of human-associated Akkermansia sp. enables access to additional growth substrates present in HMOs, including 2'-FL. Our work highlights the potential for Akkermansia to influence the development of the gut microbiota early in life and expands the known metabolic capabilities of this important human symbiont.IMPORTANCEAkkermansia are mucin-degrading specialists widely distributed in the human population. Akkermansia biwaensis has recently been observed to have enhanced growth relative to other human-associated Akkermansia on multiple human milk oligosaccharides (HMOs). However, the mechanisms for enhanced growth are not understood. Here, we characterized the phylogenetic diversity and function of select genes involved in the growth of A. biwaensis on 2'-fucosyllactose (2'-FL), a dominant HMO. Specifically, we demonstrate that two genes in a genomic locus, a putative β-galactosidase and α-fucosidase, are likely responsible for the enhanced growth on 2'-FL. The functional characterization of A. biwaensis growth on 2'-FL delineates the significance of a single genomic locus that may facilitate enhanced colonization and functional activity of select Akkermansia early in life.
Collapse
Affiliation(s)
- Loren Padilla
- Department of Biology, California State University, Northridge, California, USA
| | - Ashwana D. Fricker
- Department of Biology, California State University, Northridge, California, USA
| | - Estefani Luna
- Department of Biology, California State University, Northridge, California, USA
| | - Biswa Choudhury
- GlycoAnalytics Core, UC San Diego, Health Sciences, La Jolla, California, USA
| | - Elizabeth R. Hughes
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Maria E. Panzetta
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Raphael H. Valdivia
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Gilberto E. Flores
- Department of Biology, California State University, Northridge, California, USA
| |
Collapse
|
49
|
Liu Y, Van Horn AM, Pham MTN, Dinh BNN, Chen R, Raphael SDR, Paulino A, Thaker K, Somadder A, Frost DJ, Menke CC, Slimak ZC, Slonczewski JL. Fitness trade-offs of multidrug efflux pumps in Escherichia coli K-12 in acid or base, and with aromatic phytochemicals. Appl Environ Microbiol 2024; 90:e0209623. [PMID: 38289137 PMCID: PMC10880634 DOI: 10.1128/aem.02096-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 12/14/2023] [Indexed: 02/22/2024] Open
Abstract
Multidrug efflux pumps are the frontline defense mechanisms of Gram-negative bacteria, yet little is known of their relative fitness trade-offs under gut conditions such as low pH and the presence of antimicrobial food molecules. Low pH contributes to the proton-motive force (PMF) that drives most efflux pumps. We show how the PMF-dependent pumps AcrAB-TolC, MdtEF-TolC, and EmrAB-TolC undergo selection at low pH and in the presence of membrane-permeant phytochemicals. Competition assays were performed by flow cytometry of co-cultured Escherichia coli K-12 strains possessing or lacking a given pump complex. All three pumps showed negative selection under conditions that deplete PMF (pH 5.5 with carbonyl cyanide 3-chlorophenylhydrazone or at pH 8.0). At pH 5.5, selection against AcrAB-TolC was increased by aromatic acids, alcohols, and related phytochemicals such as methyl salicylate. The degree of fitness cost for AcrA was correlated with the phytochemical's lipophilicity (logP). Methyl salicylate and salicylamide selected strongly against AcrA, without genetic induction of drug resistance regulons. MdtEF-TolC and EmrAB-TolC each had a fitness cost at pH 5.5, but salicylate or benzoate made the fitness contribution positive. Pump fitness effects were not explained by gene expression (measured by digital PCR). Between pH 5.5 and 8.0, acrA and emrA were upregulated in the log phase, whereas mdtE expression was upregulated in the transition-to-stationary phase and at pH 5.5 in the log phase. Methyl salicylate did not affect pump gene expression. Our results suggest that lipophilic non-acidic molecules select against a major efflux pump without inducing antibiotic resistance regulons.IMPORTANCEFor drugs that are administered orally, we need to understand how ingested phytochemicals modulate drug resistance in our gut microbiome. Bacteria maintain low-level resistance by proton-motive force (PMF)-driven pumps that efflux many different antibiotics and cell waste products. These pumps play a key role in bacterial defense by conferring resistance to antimicrobial agents at first exposure while providing time for a pathogen to evolve resistance to higher levels of the antibiotic exposed. Nevertheless, efflux pumps confer energetic costs due to gene expression and pump energy expense. The bacterial PMF includes the transmembrane pH difference (ΔpH), which may be depleted by permeant acids and membrane disruptors. Understanding the fitness costs of efflux pumps may enable us to develop resistance breakers, that is, molecules that work together with antibiotics to potentiate their effect. Non-acidic aromatic molecules have the advantage that they avoid the Mar-dependent induction of regulons conferring other forms of drug resistance. We show that different pumps have distinct selection criteria, and we identified non-acidic aromatic molecules as promising candidates for drug resistance breakers.
Collapse
Affiliation(s)
- Yangyang Liu
- Department of Biology, Kenyon College, Gambier, Ohio, USA
| | | | | | | | - Rachel Chen
- Department of Biology, Kenyon College, Gambier, Ohio, USA
| | | | | | - Kavya Thaker
- Department of Biology, Kenyon College, Gambier, Ohio, USA
| | | | | | | | | | | |
Collapse
|
50
|
Tassoulas LJ, Wackett LP. Insights into the action of the pharmaceutical metformin: Targeted inhibition of the gut microbial enzyme agmatinase. iScience 2024; 27:108900. [PMID: 38318350 PMCID: PMC10839685 DOI: 10.1016/j.isci.2024.108900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 12/06/2023] [Accepted: 01/09/2024] [Indexed: 02/07/2024] Open
Abstract
Metformin is the first-line treatment for type 2 diabetes, yet its mechanism of action is not fully understood. Recent studies suggest metformin's interactions with gut microbiota are responsible for exerting therapeutic effects. In this study, we report that metformin targets the gut microbial enzyme agmatinase, as a competitive inhibitor, which may impair gut agmatine catabolism. The metformin inhibition constant (Ki) of E. coli agmatinase is 1 mM and relevant in the gut where the drug concentration is 1-10 mM. Metformin analogs phenformin, buformin, and galegine are even more potent inhibitors of E. coli agmatinase (Ki = 0.6, 0.1, and 0.007 mM, respectively) suggesting a shared mechanism. Agmatine is a known effector of human host metabolism and has been reported to augment metformin's therapeutic effects for type 2 diabetes. This gut-derived inhibition mechanism gives new insights on metformin's action in the gut and may lead to significant discoveries in improving metformin therapy.
Collapse
Affiliation(s)
- Lambros J. Tassoulas
- Department of Biochemistry, Biophysics & Molecular Biology, University of Minnesota, Minneapolis, MN 55455, USA
- BioTechnology Institute, University of Minnesota, St. Paul, MN 55108, USA
| | - Lawrence P. Wackett
- Department of Biochemistry, Biophysics & Molecular Biology, University of Minnesota, Minneapolis, MN 55455, USA
- BioTechnology Institute, University of Minnesota, St. Paul, MN 55108, USA
| |
Collapse
|