1
|
Jelescu IO, Grussu F, Ianus A, Hansen B, Barrett RLC, Aggarwal M, Michielse S, Nasrallah F, Syeda W, Wang N, Veraart J, Roebroeck A, Bagdasarian AF, Eichner C, Sepehrband F, Zimmermann J, Soustelle L, Bowman C, Tendler BC, Hertanu A, Jeurissen B, Verhoye M, Frydman L, van de Looij Y, Hike D, Dunn JF, Miller K, Landman BA, Shemesh N, Anderson A, McKinnon E, Farquharson S, Dell'Acqua F, Pierpaoli C, Drobnjak I, Leemans A, Harkins KD, Descoteaux M, Xu D, Huang H, Santin MD, Grant SC, Obenaus A, Kim GS, Wu D, Le Bihan D, Blackband SJ, Ciobanu L, Fieremans E, Bai R, Leergaard TB, Zhang J, Dyrby TB, Johnson GA, Cohen‐Adad J, Budde MD, Schilling KG. Considerations and recommendations from the ISMRM diffusion study group for preclinical diffusion MRI: Part 1: In vivo small-animal imaging. Magn Reson Med 2025; 93:2507-2534. [PMID: 40008568 PMCID: PMC11971505 DOI: 10.1002/mrm.30429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 12/19/2024] [Accepted: 12/26/2024] [Indexed: 02/27/2025]
Abstract
Small-animal diffusion MRI (dMRI) has been used for methodological development and validation, characterizing the biological basis of diffusion phenomena, and comparative anatomy. The steps from animal setup and monitoring, to acquisition, analysis, and interpretation are complex, with many decisions that may ultimately affect what questions can be answered using the resultant data. This work aims to present selected considerations and recommendations from the diffusion community on best practices for preclinical dMRI of in vivo animals. We describe the general considerations and foundational knowledge that must be considered when designing experiments. We briefly describe differences in animal species and disease models and discuss why some may be more or less appropriate for different studies. We, then, give recommendations for in vivo acquisition protocols, including decisions on hardware, animal preparation, and imaging sequences, followed by advice for data processing including preprocessing, model-fitting, and tractography. Finally, we provide an online resource that lists publicly available preclinical dMRI datasets and software packages to promote responsible and reproducible research. In each section, we attempt to provide guides and recommendations, but also highlight areas for which no guidelines exist (and why), and where future work should focus. Although we mainly cover the central nervous system (on which most preclinical dMRI studies are focused), we also provide, where possible and applicable, recommendations for other organs of interest. An overarching goal is to enhance the rigor and reproducibility of small animal dMRI acquisitions and analyses, and thereby advance biomedical knowledge.
Collapse
Affiliation(s)
- Ileana O. Jelescu
- Department of RadiologyLausanne University Hospital and University of LausanneLausanneSwitzerland
- CIBM Center for Biomedical ImagingEcole Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Francesco Grussu
- Radiomics GroupVall d'Hebron Institute of Oncology, Vall d'Hebron Barcelona Hospital CampusBarcelonaSpain
- Queen Square MS Centre, Queen Square Institute of Neurology, Faculty of Brain SciencesUniversity College LondonLondonUK
| | - Andrada Ianus
- Champalimaud ResearchChampalimaud FoundationLisbonPortugal
- School of Biomedical Engineering and Imaging SciencesKing's College LondonLondonUK
| | - Brian Hansen
- Center of Functionally Integrative NeuroscienceAarhus UniversityAarhusDenmark
| | - Rachel L. C. Barrett
- Department of NeuroimagingInstitute of Psychiatry, Psychology and Neuroscience, King's College LondonLondonUK
- NatBrainLab, Department of Forensics and Neurodevelopmental SciencesInstitute of Psychiatry, Psychology and Neuroscience, King's College LondonLondonUK
| | - Manisha Aggarwal
- Russell H. Morgan Department of Radiology and Radiological ScienceJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Stijn Michielse
- Department of Neurosurgery, School for Mental Health and Neuroscience (MHeNS)Maastricht University Medical CenterMaastrichtThe Netherlands
| | - Fatima Nasrallah
- The Queensland Brain InstituteThe University of QueenslandSt LuciaQueenslandAustralia
| | - Warda Syeda
- Melbourne Neuropsychiatry CentreThe University of MelbourneParkvilleVictoriaAustralia
| | - Nian Wang
- Department of Radiology and Imaging SciencesIndiana UniversityBloomingtonIndianaUSA
- Stark Neurosciences Research InstituteIndiana University School of MedicineBloomingtonIndianaUSA
| | - Jelle Veraart
- Center for Biomedical ImagingNYU Grossman School of MedicineNew YorkNew YorkUSA
| | - Alard Roebroeck
- Faculty of psychology and NeuroscienceMaastricht UniversityMaastrichtThe Netherlands
| | - Andrew F. Bagdasarian
- Department of Chemical and Biomedical Engineering, FAMU‐FSU College of EngineeringFlorida State UniversityTallahasseeFloridaUSA
- Center for Interdisciplinary Magnetic ResonanceNational HIgh Magnetic Field LaboratoryTallahasseeFloridaUSA
| | - Cornelius Eichner
- Department of NeuropsychologyMax Planck Institute for Human Cognitive and Brain SciencesLeipzigGermany
| | - Farshid Sepehrband
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USCUniversity of Southern CaliforniaCaliforniaLos AngelesUSA
| | - Jan Zimmermann
- Department of Neuroscience, Center for Magnetic Resonance ResearchUniversity of MinnesotaMinneapolisMinnesotaUSA
| | | | - Christien Bowman
- Bio‐Imaging Lab, Faculty of Pharmaceutical, Biomedical and Veterinary SciencesUniversity of AntwerpAntwerpBelgium
- μNEURO Research Centre of ExcellenceUniversity of AntwerpAntwerpBelgium
| | - Benjamin C. Tendler
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Andreea Hertanu
- Department of RadiologyLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Ben Jeurissen
- imec Vision Lab, Department of PhysicsUniversity of AntwerpAntwerpenBelgium
- Lab for Equilibrium Investigations and Aerospace, Department of PhysicsUniversity of AntwerpAntwerpenBelgium
| | - Marleen Verhoye
- Bio‐Imaging Lab, Faculty of Pharmaceutical, Biomedical and Veterinary SciencesUniversity of AntwerpAntwerpBelgium
- μNEURO Research Centre of ExcellenceUniversity of AntwerpAntwerpBelgium
| | - Lucio Frydman
- Department of Chemical and Biological PhysicsWeizmann Institute of ScienceRehovotIsrael
| | - Yohan van de Looij
- Division of Child Development and Growth, Department of Pediatrics, Gynaecology and Obstetrics, School of MedicineUniversité de GenèveGenèveSwitzerland
| | - David Hike
- Department of Chemical and Biomedical Engineering, FAMU‐FSU College of EngineeringFlorida State UniversityTallahasseeFloridaUSA
- Center for Interdisciplinary Magnetic ResonanceNational HIgh Magnetic Field LaboratoryTallahasseeFloridaUSA
| | - Jeff F. Dunn
- Department of Radiology, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
- Hotchkiss Brain Institute, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
- Alberta Children's Hospital Research Institute, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
| | - Karla Miller
- FMRIB Centre, Wellcome Centre for Integrative Neuroimaging, Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Bennett A. Landman
- Department of Electrical and Computer EngineeringVanderbilt UniversityNashvilleTennesseeUSA
| | - Noam Shemesh
- Champalimaud ResearchChampalimaud FoundationLisbonPortugal
| | - Adam Anderson
- Vanderbilt University Institute of Imaging ScienceVanderbilt UniversityNashvilleTennesseeUSA
- Department of Radiology and Radiological SciencesVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Emilie McKinnon
- Medical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Shawna Farquharson
- National Imaging FacilityThe University of QueenslandBrisbaneQueenslandAustralia
| | - Flavio Dell'Acqua
- Department of Forensic and Neurodevelopmental SciencesKing's College LondonLondonUK
| | - Carlo Pierpaoli
- Laboratory on Quantitative Medical imaging, NIBIBNational Institutes of HealthBethesdaMarylandUSA
| | - Ivana Drobnjak
- Department of Computer ScienceUniversity College LondonLondonUK
| | - Alexander Leemans
- PROVIDI Lab, Image Sciences InstituteUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Kevin D. Harkins
- Vanderbilt University Institute of Imaging ScienceVanderbilt UniversityNashvilleTennesseeUSA
- Radiology and Radiological SciencesVanderbilt University Medical CenterNashvilleTennesseeUSA
- Biomedical EngineeringVanderbilt UniversityNashvilleTennesseeUSA
| | - Maxime Descoteaux
- Sherbrooke Connectivity Imaing Lab (SCIL), Computer Science DepartmentUniversité de SherbrookeSherbrookeQuebecCanada
- Imeka SolutionsSherbrookeQuebecCanada
| | - Duan Xu
- Department of Radiology and Biomedical ImagingUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Hao Huang
- Department of Radiology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of RadiologyChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Mathieu D. Santin
- Centre for NeuroImaging Research (CENIR), Inserm U 1127, CNRS UMR 7225Sorbonne UniversitéParisFrance
- Paris Brain InstituteParisFrance
| | - Samuel C. Grant
- Department of Chemical and Biomedical Engineering, FAMU‐FSU College of EngineeringFlorida State UniversityTallahasseeFloridaUSA
- Center for Interdisciplinary Magnetic ResonanceNational HIgh Magnetic Field LaboratoryTallahasseeFloridaUSA
| | - Andre Obenaus
- Division of Biomedical SciencesUniversity of California RiversideRiversideCaliforniaUSA
- Preclinical and Translational Imaging CenterUniversity of California IrvineIrvineCaliforniaUSA
| | - Gene S. Kim
- Department of RadiologyWeill Cornell Medical CollegeNew YorkNew YorkUSA
| | - Dan Wu
- Key Laboratory for Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument ScienceZhejiang UniversityHangzhouChina
| | - Denis Le Bihan
- CEA, DRF, JOLIOT, NeuroSpinGif‐sur‐YvetteFrance
- Université Paris‐SaclayGif‐sur‐YvetteFrance
| | - Stephen J. Blackband
- Department of NeuroscienceUniversity of FloridaGainesvilleFloridaUSA
- McKnight Brain InstituteUniversity of FloridaGainesvilleFloridaUSA
- National High Magnetic Field LaboratoryTallahasseeFloridaUSA
| | - Luisa Ciobanu
- NeuroSpin, UMR CEA/CNRS 9027Paris‐Saclay UniversityGif‐sur‐YvetteFrance
| | - Els Fieremans
- Department of RadiologyNew York University Grossman School of MedicineNew YorkNew YorkUSA
| | - Ruiliang Bai
- Interdisciplinary Institute of Neuroscience and Technology, School of MedicineZhejiang UniversityHangzhouChina
- Frontier Center of Brain Science and Brain‐Machine IntegrationZhejiang UniversityZhejiangChina
| | - Trygve B. Leergaard
- Department of Molecular Biology, Institute of Basic Medical SciencesUniversity of OsloOsloNorway
| | - Jiangyang Zhang
- Department of RadiologyNew York University School of MedicineNew YorkNew YorkUSA
| | - Tim B. Dyrby
- Danish Research Centre for Magnetic ResonanceCentre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Amager and HvidovreHvidovreDenmark
- Department of Applied Mathematics and Computer ScienceTechnical University of DenmarkKongens LyngbyDenmark
| | - G. Allan Johnson
- Duke Center for In Vivo Microscopy, Department of RadiologyDuke UniversityDurhamNorth CarolinaUSA
- Department of Biomedical EngineeringDuke UniversityDurhamNorth CarolinaUSA
| | - Julien Cohen‐Adad
- NeuroPoly Lab, Institute of Biomedical EngineeringPolytechnique MontrealMontrealQuebecCanada
- Functional Neuroimaging Unit, CRIUGMUniversity of MontrealMontrealQuebecCanada
- Mila ‐ Quebec AI InstituteMontrealQuebecCanada
| | - Matthew D. Budde
- Department of NeurosurgeryMedical College of WisconsinMilwaukeeWisconsinUSA
- Clement J Zablocki VA Medical CenterMilwaukeeWisconsinUSA
| | - Kurt G. Schilling
- Vanderbilt University Institute of Imaging ScienceVanderbilt UniversityNashvilleTennesseeUSA
- Radiology and Radiological SciencesVanderbilt University Medical CenterNashvilleTennesseeUSA
| |
Collapse
|
2
|
Lee CY, Kuo TC, Chou YH, Peng SJ, Hsiao FT, Chung MH, Lo LW, Shen CN, Chien HJ, Chang HP, Chen CC, Jeng YM, Tien YW, Tang SC. 3D Imaging Resolves Human Pancreatic Duct-β-Cell Clusters During Cystic Change. Diabetes 2025; 74:734-748. [PMID: 39787388 PMCID: PMC12015146 DOI: 10.2337/db24-0824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
Pancreatic cystic changes in adults are increasingly identified through advanced cross-sectional imaging. However, the impact of initial/intralobular epithelial remodeling on the local β-cell population remains unclear. In this study, we examined 10 human cadaveric donor pancreases (tail and body regions) via integration of stereomicroscopy, clinical hematoxylin and eosin histology, and three-dimensional (3D) immunohistochemistry, identifying 36 microcysts (size: 1.22 ± 0.56 mm) alongside 54 low-grade pancreatic intraepithelial neoplasias (positive control of epithelial remodeling; size: 2.42 ± 1.05 mm). Both conditions exhibited significant increases in cytokeratin 7 (CK7) and insulin immunoreactive signals compared with normal lobules. Importantly, despite luminal contents of microcysts causing false positives (autofluorescence) in fluorescence imaging, the defined cystic epithelium showed distinct duct-β-cell associations-including β-cells in the epithelium and duct-β-cell clusters-visualized via antifade 3D/Airyscan superresolution imaging in the high-refractive-index polymer. The periluminal β-cells displayed insulin-positive vesicles residing near the basal domain, while the CK7+ cytokeratins in duct cells accumulated in the apical domain, underlining polarized tissue and cellular organizations. Overall, in microcyst formation, we demonstrate local and associated pancreatic exocrine and endocrine tissue remodeling. Because artifacts are a concern in β-cell investigations in a novel environment, our work using 3D-labeled human pancreas with cytokeratin and vesicle resolving powers provides a robust approach for characterizing the duct-β-cell association in a clinically relevant setting. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Chih-Yuan Lee
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Ting-Chun Kuo
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Ya-Hsien Chou
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
- Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Shih-Jung Peng
- Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Fu-Ting Hsiao
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Mei-Hsin Chung
- Department of Pathology, National Taiwan University Hospital—Hsinchu Branch, Hsinchu, Taiwan
| | - Li-Wen Lo
- Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| | - Chia-Ning Shen
- Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Hung-Jen Chien
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Hsiu-Pi Chang
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Chien-Chia Chen
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Yung-Ming Jeng
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| | - Yu-Wen Tien
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Shiue-Cheng Tang
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
- Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
3
|
Truong N, Zahra A, Lintao RCV, Chauhan R, Bento GF, Vidal Jr. M, Kim S, Lam PY, Conrads T, Conrads K, Han A, Menon R, Richardson LS. Modeling reproductive and pregnancy-associated tissues using organ-on-chip platforms: challenges, limitations, and the high throughput data frontier. Front Bioeng Biotechnol 2025; 13:1568389. [PMID: 40236940 PMCID: PMC11996799 DOI: 10.3389/fbioe.2025.1568389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 03/14/2025] [Indexed: 04/17/2025] Open
Abstract
Over the past decade, organ-on-chip technology (microphysiological systems or tissue chips) has reshaped in-vitro physiological and pathological modeling and pharmaceutical drug assessment. FDA Modernization Act 2.0 allows for alternatives to animal testing or the use of appropriate non-animal models/new approach methods (NAMs), such as Organ-on-chips (OC) platforms or in silico simulation models, to generate pre-clinical in-vitro drug trial data for regulatory purposes primes the microfluidic field to have exponential growth in the coming years. The changes in the approaches of regulatory agencies could significantly impact the development of therapeutics for use during pregnancy. However, limitations of the devices and molecular and biochemical assay shortfalls hinder the progress of the OOC field. This review describes available reproductive and pregnancy-related OOC platforms, and the current methodologies utilized to generate endpoint datasets (e.g., microscopic imaging, immunocytochemistry, real-time polymerase chain reaction, cytokine multiplex analysis). Microfluidic platform limitations, such as fewer number of cells or low supernatant volumes and restrictions regarding fabrication materials, are described. Novel approaches (e.g., spatial transcriptomics, imaging cytometry by time of flight, exosomes analysis using Exoview) to overcome these challenges are described. OOC platforms are primed to provide biologically relevant and clinically translational data that can revolutionize in-vitro physiological modeling, drug discovery, and toxicologic risk assessment. However, engineering adaptations to increase the throughput of devices (i.e., device arrays) and biological advancements to improve data throughput are both needed for these platforms to reach their full potential.
Collapse
Affiliation(s)
- Nina Truong
- John Sealy School of Medicine, University Blvd., Galveston, TX, United States
| | - Abir Zahra
- John Sealy School of Medicine, University Blvd., Galveston, TX, United States
| | - Ryan C. V. Lintao
- Institute of Reproductive Health, National Institutes of Health, University of the Philippines Manila, Manila, Philippines
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Rahul Chauhan
- John Sealy School of Medicine, University Blvd., Galveston, TX, United States
| | - Giovana Fernanda Bento
- Department of Pathology, Botucatu Medical School, São Paulo State University, São Paulo, Brazil
| | - Manuel Vidal Jr.
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
- College of Medicine, San Beda University, Manila, Philippines
- Department of Chemistry, College of Science, De La Salle University Manila, Manila, Philippines
| | - Sungjin Kim
- Department of Biomedical Engineering and Electrical and Computer Engineering, Texas A&M University, College Station, TX, United States
| | - Po Yi Lam
- Department of Biomedical Engineering and Electrical and Computer Engineering, Texas A&M University, College Station, TX, United States
| | - Thomas Conrads
- Gynecologic Cancer Center of Excellence, Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Women’s Health Integrated Research Center, Women’s Service Line, Inova Health System, Falls Church, VA, United States
| | - Kelly Conrads
- Gynecologic Cancer Center of Excellence, Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Women’s Health Integrated Research Center, Women’s Service Line, Inova Health System, Falls Church, VA, United States
| | - Arum Han
- Department of Biomedical Engineering and Electrical and Computer Engineering, Texas A&M University, College Station, TX, United States
| | - Ramkumar Menon
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Lauren S. Richardson
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| |
Collapse
|
4
|
Collins J, Farnsworth NL. Active targeting of type 1 diabetes therapies to pancreatic beta cells using nanocarriers. Diabetologia 2025; 68:692-703. [PMID: 39847085 DOI: 10.1007/s00125-024-06356-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 11/21/2024] [Indexed: 01/24/2025]
Abstract
Type 1 diabetes is an autoimmune disease characterised by the destruction of pancreatic beta cells, resulting in lifelong insulin dependence. Although exogenous insulin can maintain glycaemic control, this approach does not protect residual or replacement pancreatic beta cells from immune-mediated death. Current therapeutics designed to protect functional beta cell mass or promote beta cell proliferation and regeneration can have off-target effects, resulting in higher dose requirements and adverse side effects. Targeted drug delivery using nanocarriers has demonstrated potential for overcoming these limitations. The critical bottleneck limiting the development of beta cell-targeted therapies is a lack of highly specific beta cell markers. This review provides an overview of the use of nanocarriers for cell-targeted delivery and the current state of the field of beta cell targeting. Technologies such as systematic evolution of ligands by exponential enrichment (SELEX) aptamer selection, phage display screening, and omics datasets from human samples are highlighted as tools to identify novel beta cell-specific targets that can be combined with nanocarriers for targeted delivery of therapeutics. Ultimately, beta cell-targeted therapies using nanocarriers present a unique opportunity to develop tailored treatments for each stage of type 1 diabetes with the goal of providing individuals with treatment options that prevent further progression or reverse the course of the disease.
Collapse
Affiliation(s)
- Jillian Collins
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, CO, USA
| | - Nikki L Farnsworth
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, CO, USA.
| |
Collapse
|
5
|
Hao Y, Horak J, Stijepic Z, Can SN, Tu L, Wolff JA, Koletzko B. Comprehensive tissue homogenization and metabolite extraction for application in clinical metabolomics. Anal Chim Acta 2025; 1344:343728. [PMID: 39984215 DOI: 10.1016/j.aca.2025.343728] [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: 10/18/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/23/2025]
Abstract
BACKGROUND Metabolomics and lipidomics analysis of various biological samples offer insights into potential mechanisms of health and disease development. Tissue samples, compared to other biological samples, are less elucidated due to challenges in sample collection and lack of standardized sample preparation protocols for reproducible tissue homogenization and broad-range metabolite extraction. RESULTS Pork tissue samples were homogenized with six different solvent mixtures with increasing lipophilicity, followed by metabolites extraction using methanol for polar and methyl-tert-butyl ether (MTBE) in methanol (MeOH) for highly lipophilic compounds. Metabolite profiles of supernatant and homogenate extraction for three extract volumes were compared. Solvent dependent pipette tip blockage was addressed by introduction of a prewetting correction factor for non-polar homogenization solutions and low volume tissue homogenate pipetting. Upset plots were applied for multi-dimensional metabolite extraction efficiency evaluation for 24 different sample preparation conditions. The best-performing homogenization solution was PBS; MeOH (1:1; v/v), combined with a two-step polar metabolite and lipid extraction using MeOH and 75 % MTBE in MeOH employing the tissue homogenate. The optimized experimental conditions were applied on mouse pancreas tissues, providing evidence of varying metabolic pathway activities across different anatomical regions of an organ. SIGNIFICANCE This study introduces a comprehensive tissue sample preparation and metabolite quantification workflow, covering highly polar to highly lipophilic metabolites using targeted high performance liquid chromatography electrospray ionization triple quadrupole-linear ion trap mass spectrometer (HPLC-ESI-QTRAP-MS/MS) for absolute quantitation of amino acids, organic acids and keto-acids, acyl-carnitines, and phospho-choline lipids.
Collapse
Affiliation(s)
- Yuntao Hao
- Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital, LMU Munich, Lindwurmstrasse 4, D-80337, Munich, Germany; German Center for Child and Adolescent Health (DZKJ), site, Munich, Germany
| | - Jeannie Horak
- Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital, LMU Munich, Lindwurmstrasse 4, D-80337, Munich, Germany; German Center for Child and Adolescent Health (DZKJ), site, Munich, Germany.
| | - Zorica Stijepic
- Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital, LMU Munich, Lindwurmstrasse 4, D-80337, Munich, Germany; German Center for Child and Adolescent Health (DZKJ), site, Munich, Germany
| | - Sultan Nilay Can
- Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital, LMU Munich, Lindwurmstrasse 4, D-80337, Munich, Germany; German Center for Child and Adolescent Health (DZKJ), site, Munich, Germany
| | - Luke Tu
- Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital, LMU Munich, Lindwurmstrasse 4, D-80337, Munich, Germany; German Center for Child and Adolescent Health (DZKJ), site, Munich, Germany
| | - Julia Alexandra Wolff
- Department of Medicine II, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
| | - Berthold Koletzko
- Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital, LMU Munich, Lindwurmstrasse 4, D-80337, Munich, Germany; German Center for Child and Adolescent Health (DZKJ), site, Munich, Germany.
| |
Collapse
|
6
|
Kwon Y, Yoon H, Ha J, Lee HS, Pahk K, Kwon H, Kim S, Park S. Changes in pancreatic levodopa uptake in patients with obesity and new-onset type 2 diabetes: an 18F-FDOPA PET-CT study. Front Endocrinol (Lausanne) 2025; 16:1460253. [PMID: 40099262 PMCID: PMC11911206 DOI: 10.3389/fendo.2025.1460253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 01/23/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction Levodopa (L-3,4-dihydroxyphenylalanine)g, a dopamine precursor that circulates in the peripheral region, is involved in pancreatic glycemic control. Although previous animal studies have shown that peripheral levodopa is correlated with insulin secretion in pancreatic beta cells, the mechanism by which the pancreas uses levodopa differently in humans with obesity and type 2 diabetes remains unknown. Our study aimed to observe how the pancreas uptakes and utilizes levodopa differently under obese and diabetic conditions. Materials and method 18F-fluoro-L-dopa positron emission tomography-computed tomography (18F-FDOPA PET-CT) was used to visualize how the human body uses levodopa under obese and diabetic conditions and presented its clinical implications. 10 patients were divided into 3 groups: 1) Group A, normal weight without type 2 diabetes; 2) Group B, obese without type 2 diabetes; and 3) Group C, obese with new-onset type 2 diabetes. All patients' lifestyle modification was conducted prior to 18F-FDOPA PET-CT, and plasma samples were collected to confirm changes in amino acid metabolites. Results Pancreatic levodopa uptake increased in obese patients with insulin resistance, whereas it decreased in obese patients with new-onset type 2 diabetes [standardized uptake value (SUV) mean in participants with normal weight, 2.6 ± 0.7; SUVmean in patients with obesity, 3.6 ± 0.1; SUVmean in patients with obesity and new-onset type 2 diabetes, 2.6 ± 0.1, P = 0.02]. Conclusions This suggested that the alterations in the functional capacity of pancreatic beta cells to take up circulating levodopa are potentially linked to the insulin resistance and the pathogenesis of type 2 diabetes. The differences in the uptake values between the groups implied that pancreatic levodopa uptake could be an early indicator of type 2 diabetes.
Collapse
Affiliation(s)
- Yeongkeun Kwon
- Center for Obesity and Metabolic Diseases, Korea University Anam Hospital, Seoul, Republic of Korea
- Gut & Metabolism Laboratory, Korea University College of Medicine, Seoul, Republic of Korea
- Division of Foregut Surgery, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hanseok Yoon
- Division of Biotechnology, Korea University, Seoul, Republic of Korea
| | - Jane Ha
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, United States
| | - Hyeon-Seong Lee
- Gangneung Institute of Natural Products, Korea Institute of Science and Technology, Gangneung, Republic of Korea
| | - Kisoo Pahk
- Department of Nuclear Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hyunwoo Kwon
- Department of Nuclear Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Sungeun Kim
- Department of Nuclear Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Sungsoo Park
- Center for Obesity and Metabolic Diseases, Korea University Anam Hospital, Seoul, Republic of Korea
- Gut & Metabolism Laboratory, Korea University College of Medicine, Seoul, Republic of Korea
- Division of Foregut Surgery, Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
7
|
Hilliard BK, Prendergast JE, Smith MJ. Dia-B-Ties: B Cells in the Islet-Immune-Cell Interface in T1D. Biomolecules 2025; 15:332. [PMID: 40149868 PMCID: PMC11940010 DOI: 10.3390/biom15030332] [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: 02/08/2025] [Revised: 02/20/2025] [Accepted: 02/22/2025] [Indexed: 03/29/2025] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease that affects an estimated 30 million people worldwide and results in a lifelong dependency of exogenous insulin treatments. While T1D is characterized by T-cell driven-destruction of the insulin-secreting β cells, B lymphocytes play a key role in the islet-immune interface. B cells are an essential intermediary between islet cells and other immune-cell populations. Through antigen presentation, cytokine secretion, and antibody production, B cells play a role in activating autoreactive islet-specific T cells, thus potentiating pancreatic inflammation in the early stages of T1D. Despite this, their role in disease development remains an understudied feature of T1D with significant therapeutic potential. Herein, we will discuss the current knowledge of the islet-immune-cell interface within T1D through the lens of B lymphocytes. We will also consider knowledge gaps that may be limiting further therapeutic opportunities.
Collapse
Affiliation(s)
- Brandon K. Hilliard
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO 80045, USA
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Jessica E. Prendergast
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO 80045, USA
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Mia J. Smith
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO 80045, USA
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| |
Collapse
|
8
|
Gravdal A, Wilhelm SJ, Lowe ME, Molven A, Xiao XK, Fjeld K. The MODY-Causing Mutation of the Human Carboxyl Ester Lipase Gene (CEL) Triggers Chronic Pancreatitis but not Diabetes in Mice. Gastroenterology 2025:S0016-5085(25)00367-1. [PMID: 39956334 DOI: 10.1053/j.gastro.2025.01.243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 01/02/2025] [Accepted: 01/22/2025] [Indexed: 02/18/2025]
Affiliation(s)
- Anny Gravdal
- The Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, Bergen, Norway; Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Steven J Wilhelm
- Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri
| | - Mark E Lowe
- Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri
| | - Anders Molven
- The Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, Bergen, Norway; Department of Pathology and Section for Cancer Genomics, Laboratory Clinic, Bergen, Norway; Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Xunjun K Xiao
- Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri
| | - Karianne Fjeld
- The Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, Bergen, Norway; Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway.
| |
Collapse
|
9
|
Paradiž Leitgeb E, Pohorec V, Križančić Bombek L, Skelin Klemen M, Duh M, Gosak M, Dolenšek J, Stožer A. Calcium Imaging and Analysis in Beta Cells in Acute Mouse Pancreas Tissue Slices. Methods Mol Biol 2025; 2861:223-246. [PMID: 39395109 DOI: 10.1007/978-1-0716-4164-4_17] [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: 10/14/2024]
Abstract
Ca2+ ions play a central role in the stimulus-secretion coupling cascade of pancreatic beta cells. The use of confocal microscopy in conjunction with the acute pancreas tissue slice technique offers valuable insights into changes in the intracellular calcium concentration following stimulation by secretagogues. This allows the study of beta cells on a single cell level, as well as their behavior on a multicellular scale within an intact environment. With the use of advanced analytical tools, this approach offers insight into how single cells contribute to the functional unit of islets of Langerhans and processes underlying insulin secretion. Here we describe a comprehensive protocol for the preparation and utilization of acute pancreas tissue slices in mice, the use of high-resolution confocal microscopy for observation of glucose-stimulated calcium dynamics in beta cells, and the computational analysis for objective evaluation of calcium signals.
Collapse
Affiliation(s)
| | - Viljem Pohorec
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | | | | | - Maja Duh
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
| | - Marko Gosak
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
- Alma Mater Europaea University, Maribor, Slovenia
| | - Jurij Dolenšek
- Faculty of Medicine, University of Maribor, Maribor, Slovenia.
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia.
| | - Andraž Stožer
- Faculty of Medicine, University of Maribor, Maribor, Slovenia.
| |
Collapse
|
10
|
Hahm J, Kumar D, Andrade JAF, Arany E, Hill DJ. Bi-Hormonal Endocrine Cell Presence Within the Islets of Langerhans of the Human Pancreas Throughout Life. Cells 2025; 14:34. [PMID: 39791735 PMCID: PMC11719505 DOI: 10.3390/cells14010034] [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: 11/28/2024] [Revised: 12/20/2024] [Accepted: 12/29/2024] [Indexed: 01/12/2025] Open
Abstract
Bi-hormonal islet endocrine cells have been proposed to represent an intermediate state of cellular transdifferentiation, enabling an increase in beta-cell mass in response to severe metabolic stress. Beta-cell plasticity and regenerative capacity are thought to decrease with age. We investigated the ontogeny of bi-hormonal islet endocrine cell populations throughout the human lifespan. Immunofluorescence microscopy was performed for insulin, glucagon, and somatostatin presence on paraffin-embedded sections of pancreata from 20 donors without diabetes aged between 11 days and 79 years of age. The mean proportional presence of glucagon-, insulin-, and somatostatin-immunoreactive cells within islets was 27.5%, 62.1%, and 12.1%, respectively. There was no change in the relative presence of alpha- or beta-cells with advancing age, but delta-cell presence showed a decline with age (R2 = 0.59, p < 0.001). The most abundant bi-hormonal cell phenotype observed co-stained for glucagon and insulin, representing 3.1 ± 0.3% of all islet cells. Glucagon/somatostatin and insulin/somatostatin bi-hormonal cells were also observed representing 2-3% abundance relative to islet cell number. Glucagon/insulin bi-hormonal cells increased with age (R2 = 0.30, p < 0.05) whilst insulin/somatostatin (R2 = 0.50, p < 0.01) and glucagon/somatostatin (R2 = 0.35, p < 0.05) cells decreased with age of donor. Findings show that bi-hormonal cells are present within human pancreatic islets throughout life, perhaps reflecting an ongoing potential for endocrine cell plasticity.
Collapse
Affiliation(s)
- Jiwon Hahm
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada; (J.H.); (J.A.F.A.)
- Lawson Health Research Institute, St. Joseph’s Health Care, London, ON N6A 4V2, Canada; (D.K.); (E.A.)
| | - Dawn Kumar
- Lawson Health Research Institute, St. Joseph’s Health Care, London, ON N6A 4V2, Canada; (D.K.); (E.A.)
- Faculty of Science, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Juan Andres Fernandez Andrade
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada; (J.H.); (J.A.F.A.)
- Lawson Health Research Institute, St. Joseph’s Health Care, London, ON N6A 4V2, Canada; (D.K.); (E.A.)
| | - Edith Arany
- Lawson Health Research Institute, St. Joseph’s Health Care, London, ON N6A 4V2, Canada; (D.K.); (E.A.)
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
| | - David J. Hill
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada; (J.H.); (J.A.F.A.)
- Lawson Health Research Institute, St. Joseph’s Health Care, London, ON N6A 4V2, Canada; (D.K.); (E.A.)
- Faculty of Science, McMaster University, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
11
|
Aplin AC, Aghazadeh Y, Mohn OG, Hull-Meichle RL. Role of the Pancreatic Islet Microvasculature in Health and Disease. J Histochem Cytochem 2024; 72:711-728. [PMID: 39601198 PMCID: PMC11600425 DOI: 10.1369/00221554241299862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
The pancreatic islet vasculature comprises microvascular endothelial cells surrounded by mural cells (pericytes). Both cell types support the islet by providing (1) a conduit for delivery and exchange of nutrients and hormones; (2) paracrine signals and extracellular matrix (ECM) components that support islet development, architecture, and endocrine function; and (3) a barrier against inflammation and immune cell infiltration. In type 2 diabetes, the islet vasculature becomes inflamed, showing loss of endothelial cells, detachment, and/or trans-differentiation of pericytes, vessel dilation, and excessive ECM deposition. While most work to date has focused either on endothelial cells or pericytes in isolation, it is very likely that the interaction between these cell types and disruption of that interaction in diabetes are critically important. In fact, dissociation of pericytes from endothelial cells is an early, key feature of microvascular disease in multiple tissues/disease states. Moreover, in beta-cell replacement therapy, co-transplantation with microvessels versus endothelial cells alone is substantially more effective in improving survival and function of the transplanted cells. Ongoing studies, including characterization of islet vascular cell signatures, will aid in the identification of new therapeutic targets aimed at improving islet function and benefiting people living with all forms of diabetes.
Collapse
Affiliation(s)
- Alfred C. Aplin
- Seattle Institute for Biomedical and Clinical Research, and Research Service, Department of Veterans Affairs Puget Sound Health Care System, Seattle, Washington
| | - Yasaman Aghazadeh
- Institut de Recherches Cliniques de Montreal (IRCM), Department of Medicine, University of Montreal, and Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
| | - Olivia G. Mohn
- Seattle Institute for Biomedical and Clinical Research, and Research Service, Department of Veterans Affairs Puget Sound Health Care System, Seattle, Washington
| | - Rebecca L. Hull-Meichle
- Seattle Institute for Biomedical and Clinical Research, and Research Service, Department of Veterans Affairs Puget Sound Health Care System, Seattle, Washington
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, Washington; and Alberta Diabetes Institute and Department of Cell Biology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
12
|
Venglovecz V, Grassalkovich A, Tóth E, Ébert A, Gál E, Korsós MM, Maléth J, Rakonczay Z, Galla Z, Monostori P, Hegyi P. Restoring CFTR function with Orkambi decreases the severity of alcohol-induced acute pancreatitis. J Physiol 2024; 602:6153-6170. [PMID: 39418107 DOI: 10.1113/jp287289] [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: 07/11/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024] Open
Abstract
Heavy alcohol intake is one of the most common causes of acute pancreatitis (AP). We have previously shown that ethanol (EtOH) decreases the expression and activity of the cystic fibrosis transmembrane conductance regulator (CFTR), which plays a key role in alcohol-induced AP development. The prescription drug, Orkambi (a combination of ivacaftor and lumacaftor) can correct impaired CFTR function and expression in cystic fibrosis (CF) patients. Thus, the present study aimed to investigate whether Orkambi can mitigate alcohol-induced AP. Intact guinea-pig pancreatic ducts were pre-treated with different concentrations of ethanol (EtOH; 30, 50 and 100 mm) for 12 h alone or in combination with ivacaftor (VX770) and/or lumacaftor (VX-809), and CFTR expression and activity were evaluated by immunostaining and by the patch clamp technique, respectively. Alcoholic AP was induced in Orkambi-treated guinea-pigs, and standard laboratory and histological parameters were measured. Ivacaftor and lumacaftor alone or in combination dose-dependently restored the apical expression and activity of CFTR after EtOH treatment in vitro. Oral administration of Orkambi reduced the severity of alcohol-induced AP and restored impaired CFTR activity and expression. Orkambi is able to restore the CFTR defect caused by EtOH and decreases the severity of alcohol-induced pancreatitis. This is the first in vivo pre-clinical evidence of Orkambi efficacy in the treatment of alcohol-induced AP. KEY POINTS: Acute pancreatitis is one of the leading causes of hospital admission among gastrointestinal diseases in which the lack of a specific drug therapy plays a crucial role. The cystic fibrosis transmembrane conductance regulator (CFTR) plays an essential role in pancreatic ductal HCO3 - secretion; inappropriate CFTR function, as seen in heavy alcohol consumption, increases the risk of pancreatitis development. CFTR modulators are able to prevent the inhibitory effect of ethanol and reduce pancreatic ductal injury and the severity of alcohol-induced pancreatitis. CFTR modulators present a novel option in the pharmacotherapy of alcohol-induced pancreatitis by enhancing pancreatic functions or preventing recurrence.
Collapse
Affiliation(s)
- Viktória Venglovecz
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
- Translational Pancreatology Research Group, Interdisciplinary Center of Excellence for Research Development and Innovation, University of Szeged, Szeged, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Anna Grassalkovich
- Translational Pancreatology Research Group, Interdisciplinary Center of Excellence for Research Development and Innovation, University of Szeged, Szeged, Hungary
- Department of Medicine, University of Szeged, Szeged, Hungary
| | - Emese Tóth
- Translational Pancreatology Research Group, Interdisciplinary Center of Excellence for Research Development and Innovation, University of Szeged, Szeged, Hungary
- Department of Medicine, University of Szeged, Szeged, Hungary
- Department of Health Sciences, Department of Theoretical and Integrative Health Sciences, University of Debrecen, Debrecen, Hungary
| | - Attila Ébert
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - Eleonóra Gál
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | | | - József Maléth
- Department of Medicine, University of Szeged, Szeged, Hungary
- HCEMM-SZTE Molecular Gastroenterology Research Group, University of Szeged, Szeged, Hungary
- ELKH-USZ Momentum Epithelial Cell Signaling and Secretion Research Group, University of Szeged, Szeged, Hungary
| | - Zoltán Rakonczay
- Department of Pathophysiology, University of Szeged, Szeged, Hungary
| | - Zsolt Galla
- Metabolic and Newborn Screening Laboratory, Department of Paediatrics, University of Szeged, Szeged, Hungary
| | - Péter Monostori
- Metabolic and Newborn Screening Laboratory, Department of Paediatrics, University of Szeged, Szeged, Hungary
| | - Péter Hegyi
- Translational Pancreatology Research Group, Interdisciplinary Center of Excellence for Research Development and Innovation, University of Szeged, Szeged, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Institute for Pancreatic Disorders, Semmelweis University, Budapest, Hungary
| |
Collapse
|
13
|
Hahm J, Thirunavukarasu B, Gadoo R, Andrade JAF, Dalton T, Arany E, Hill DJ. Alpha- to Beta-Cell Transdifferentiation in Neonatal Compared with Adult Mouse Pancreas in Response to a Modest Reduction in Beta-Cells Using Streptozotocin. Int J Mol Sci 2024; 25:11152. [PMID: 39456933 PMCID: PMC11508719 DOI: 10.3390/ijms252011152] [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: 09/20/2024] [Revised: 10/05/2024] [Accepted: 10/13/2024] [Indexed: 10/28/2024] Open
Abstract
Following the near-total depletion of pancreatic beta-cells with streptozotocin (STZ), a partial recovery of beta-cell mass (BCM) can occur, in part due to the alpha- to beta-cell transdifferentiation with an intermediary insulin/glucagon bi-hormonal cell phenotype. However, human type 2 diabetes typically involves only a partial reduction in BCM and it is not known if recovery after therapeutic intervention involves islet cell transdifferentiation, or how this varies with age. Here, we used transgenic mouse models to examine if islet cell transdifferentiation contributes to BCM recovery following only a partial depletion of BCM. Cell lineage tracing was employed using Glucagon-Cre/yellow fluorescent protein (YFP) transgenic mice treated with STZ (25 mg/kg-neonates; 70 mg/kg-adults) or vehicle alone on 3 consecutive days. Mice were euthanized 2-30 days later with a prior glucose tolerance test on day 30, and immunofluorescence histology performed on the pancreata. Beta-cell abundance was reduced by 30-40% two days post STZ in both neonates and adults, and subsequently partially recovered in adult but not neonatal mice. Glucose tolerance recovered in adult females, but not in males or neonates. Bi-hormonal cell abundance increased 2-3-fold in STZ-treated mice vs. controls in both neonates and adults, as did transdifferentiated cells expressing insulin and the YFP lineage tag, but not glucagon. Transdifferentiated cell presence was an order of magnitude lower than that of bi-hormonal cells. We conclude that alpha- to beta-cell transdifferentiation occurs in mice following only a moderate depletion in BCM, and that this was accompanied by a partial recovery of BCM in adults.
Collapse
Affiliation(s)
- Jiwon Hahm
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada; (J.H.); (B.T.); (J.A.F.A.); (T.D.)
- Lawson Health Research Institute, St. Joseph’s Health Care, London, ON N6A 4V2, Canada; (R.G.); (E.A.)
| | - Bavina Thirunavukarasu
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada; (J.H.); (B.T.); (J.A.F.A.); (T.D.)
- Lawson Health Research Institute, St. Joseph’s Health Care, London, ON N6A 4V2, Canada; (R.G.); (E.A.)
| | - Reva Gadoo
- Lawson Health Research Institute, St. Joseph’s Health Care, London, ON N6A 4V2, Canada; (R.G.); (E.A.)
- Faculty of Science, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Juan Andres Fernandez Andrade
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada; (J.H.); (B.T.); (J.A.F.A.); (T.D.)
- Lawson Health Research Institute, St. Joseph’s Health Care, London, ON N6A 4V2, Canada; (R.G.); (E.A.)
| | - Tyler Dalton
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada; (J.H.); (B.T.); (J.A.F.A.); (T.D.)
- Lawson Health Research Institute, St. Joseph’s Health Care, London, ON N6A 4V2, Canada; (R.G.); (E.A.)
| | - Edith Arany
- Lawson Health Research Institute, St. Joseph’s Health Care, London, ON N6A 4V2, Canada; (R.G.); (E.A.)
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
| | - David J. Hill
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada; (J.H.); (B.T.); (J.A.F.A.); (T.D.)
- Lawson Health Research Institute, St. Joseph’s Health Care, London, ON N6A 4V2, Canada; (R.G.); (E.A.)
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
| |
Collapse
|
14
|
Maestas MM, Bui MH, Millman JR. Recent progress in modeling and treating diabetes using stem cell-derived islets. Stem Cells Transl Med 2024; 13:949-958. [PMID: 39159002 PMCID: PMC11465181 DOI: 10.1093/stcltm/szae059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 07/12/2024] [Indexed: 08/21/2024] Open
Abstract
Stem cell-derived islets (SC-islets) offer the potential to be an unlimited source of cells for disease modeling and the treatment of diabetes. SC-islets can be genetically modified, treated with chemical compounds, or differentiated from patient derived stem cells to model diabetes. These models provide insights into disease pathogenesis and vulnerabilities that may be targeted to provide treatment. SC-islets themselves are also being investigated as a cell therapy for diabetes. However, the transplantation process is imperfect; side effects from immunosuppressant use have reduced SC-islet therapeutic potential. Alternative methods to this include encapsulation, use of immunomodulating molecules, and genetic modification of SC-islets. This review covers recent advances using SC-islets to understand different diabetes pathologies and as a cell therapy.
Collapse
Affiliation(s)
- Marlie M Maestas
- Roy and Diana Vagelos Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO 63110, United States
- Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Maggie H Bui
- Roy and Diana Vagelos Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Jeffrey R Millman
- Roy and Diana Vagelos Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO 63110, United States
- Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, MO 63110, United States
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63110, United States
| |
Collapse
|
15
|
Huang P, Zhu Y, Qin J. Research advances in understanding crosstalk between organs and pancreatic β-cell dysfunction. Diabetes Obes Metab 2024; 26:4147-4164. [PMID: 39044309 DOI: 10.1111/dom.15787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/25/2024]
Abstract
Obesity has increased dramatically worldwide. Being overweight or obese can lead to various conditions, including dyslipidaemia, hypertension, glucose intolerance and metabolic syndrome (MetS), which may further lead to type 2 diabetes mellitus (T2DM). Previous studies have identified a link between β-cell dysfunction and the severity of MetS, with multiple organs and tissues affected. Identifying the associations between pancreatic β-cell dysfunction and organs is critical. Research has focused on the interaction between the liver, gut and pancreatic β-cells. However, the mechanisms and related core targets are still not perfectly elucidated. The aims of this review were to summarize the mechanisms of β-cell dysfunction and to explore the potential pathogenic pathways and targets that connect the liver, gut, adipose tissue, muscle, and brain to pancreatic β-cell dysfunction.
Collapse
Affiliation(s)
- Peng Huang
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yunling Zhu
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jian Qin
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
16
|
Perez-Frances M, Bru-Tari E, Cohrs C, Abate MV, van Gurp L, Furuyama K, Speier S, Thorel F, Herrera PL. Regulated and adaptive in vivo insulin secretion from islets only containing β-cells. Nat Metab 2024; 6:1791-1806. [PMID: 39169271 PMCID: PMC11422169 DOI: 10.1038/s42255-024-01114-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 07/22/2024] [Indexed: 08/23/2024]
Abstract
Insulin-producing β-cells in pancreatic islets are regulated by systemic cues and, locally, by adjacent islet hormone-producing 'non-β-cells' (namely α-cells, δ-cells and γ-cells). Yet whether the non-β-cells are required for accurate insulin secretion is unclear. Here, we studied mice in which adult islets are exclusively composed of β-cells and human pseudoislets containing only primary β-cells. Mice lacking non-β-cells had optimal blood glucose regulation, enhanced glucose tolerance, insulin sensitivity and restricted body weight gain under a high-fat diet. The insulin secretion dynamics in islets composed of only β-cells was comparable to that in intact islets. Similarly, human β-cell pseudoislets retained the glucose-regulated mitochondrial respiration, insulin secretion and exendin-4 responses of entire islets. The findings indicate that non-β-cells are dispensable for blood glucose homeostasis and β-cell function. These results support efforts aimed at developing diabetes treatments by generating β-like clusters devoid of non-β-cells, such as from pluripotent stem cells differentiated in vitro or by reprograming non-β-cells into insulin producers in situ.
Collapse
Affiliation(s)
- Marta Perez-Frances
- Department of Genetic Medicine and Development, iGE3 and Centre facultaire du diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Eva Bru-Tari
- Department of Genetic Medicine and Development, iGE3 and Centre facultaire du diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Christian Cohrs
- Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, Neuherberg, Germany
| | - Maria Valentina Abate
- Department of Genetic Medicine and Development, iGE3 and Centre facultaire du diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Léon van Gurp
- Department of Genetic Medicine and Development, iGE3 and Centre facultaire du diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Kenichiro Furuyama
- Department of Genetic Medicine and Development, iGE3 and Centre facultaire du diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Stephan Speier
- Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, Neuherberg, Germany
| | - Fabrizio Thorel
- Department of Genetic Medicine and Development, iGE3 and Centre facultaire du diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Pedro L Herrera
- Department of Genetic Medicine and Development, iGE3 and Centre facultaire du diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
17
|
Lee Y, Lee K. Pancreatic Diseases: Genetics and Modeling Using Human Pluripotent Stem Cells. Int J Stem Cells 2024; 17:253-269. [PMID: 38664226 PMCID: PMC11361847 DOI: 10.15283/ijsc24036] [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: 03/29/2024] [Revised: 04/01/2024] [Accepted: 04/01/2024] [Indexed: 08/31/2024] Open
Abstract
Pancreas serves endocrine and exocrine functions in the body; thus, their pathology can cause a broad range of irreparable consequences. Endocrine functions include the production of hormones such as insulin and glucagon, while exocrine functions involve the secretion of digestive enzymes. Disruption of these functions can lead to conditions like diabetes mellitus and exocrine pancreatic insufficiency. Also, the symptoms and causality of pancreatic cancer very greatly depends on their origin: pancreatic ductal adenocarcinoma is one of the most fatal cancer; however, most of tumor derived from endocrine part of pancreas are benign. Pancreatitis, an inflammation of the pancreatic tissues, is caused by excessive alcohol consumption, the bile duct obstruction by gallstones, and the premature activation of digestive enzymes in the pancreas. Hereditary pancreatic diseases, such as maturity-onset diabetes of the young and hereditary pancreatitis, can be a candidate for disease modeling using human pluripotent stem cells (hPSCs), due to their strong genetic influence. hPSC-derived pancreatic differentiation has been established for cell replacement therapy for diabetic patients and is robustly used for disease modeling. The disease modeling platform that allows interactions between immune cells and pancreatic cells is necessary to perform in-depth investigation of disease pathogenesis.
Collapse
Affiliation(s)
- Yuri Lee
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Korea
| | - Kihyun Lee
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Korea
- College of Pharmacy, Ewha Womans University, Seoul, Korea
| |
Collapse
|
18
|
Deepa Maheshvare M, Charaborty R, Haldar S, Raha S, Pal D. Kiphynet: an online network simulation tool connecting cellular kinetics and physiological transport. Metabolomics 2024; 20:94. [PMID: 39110256 DOI: 10.1007/s11306-024-02151-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 07/10/2024] [Indexed: 10/22/2024]
Abstract
INTRODUCTION Human metabolism is sustained by functional networks that operate at diverse scales. Capturing local and global dynamics in the human body by hierarchically bridging multi-scale functional networks is a major challenge in physiological modeling. OBJECTIVES To develop an interactive, user-friendly web application that facilitates the simulation and visualization of advection-dispersion transport in three-dimensional (3D) microvascular networks, biochemical exchange, and metabolic reactions in the tissue layer surrounding the vasculature. METHODS To help modelers combine and simulate biochemical processes occurring at multiple scales, KiPhyNet deploys our discrete graph-based modeling framework that bridges functional networks existing at diverse scales. KiPhyNet is implemented in Python based on Apache web server using MATLAB as the simulator engine. KiPhyNet provides the functionality to assimilate multi-omics data from clinical and experimental studies as well as vascular data from imaging studies to investigate the role of structural changes in vascular topology on the functional response of the tissue. RESULTS With the network topology, its biophysical attributes, values of initial and boundary conditions, parameterized kinetic constants, biochemical species-specific transport properties such as diffusivity as inputs, a user can use our application to simulate and view the simulation results. The results of steady-state velocity and pressure fields and dynamic concentration fields can be interactively examined. CONCLUSION KiPhyNet provides barrier-free access to perform time-course simulation experiments by building multi-scale models of microvascular networks in physiology, using a discrete modeling framework. KiPhyNet is freely accessible at http://pallab.cds.iisc.ac.in/kiphynet/ and the documentation is available at https://deepamahm.github.io/kiphynet_docs/ .
Collapse
Affiliation(s)
- M Deepa Maheshvare
- Department of Computational and Data Sciences, Indian Institute of Science, Bangalore, 560012, India
| | - Rohit Charaborty
- Department of Computational and Data Sciences, Indian Institute of Science, Bangalore, 560012, India
| | - Subhraneel Haldar
- Department of Computational and Data Sciences, Indian Institute of Science, Bangalore, 560012, India
| | - Soumyendu Raha
- Department of Computational and Data Sciences, Indian Institute of Science, Bangalore, 560012, India
| | - Debnath Pal
- Department of Computational and Data Sciences, Indian Institute of Science, Bangalore, 560012, India.
| |
Collapse
|
19
|
Wang L, Bai Y, Cao Z, Guo Z, Lian Y, Liu P, Zeng Y, Lyu W, Chen Q. Histone deacetylases and inhibitors in diabetes mellitus and its complications. Biomed Pharmacother 2024; 177:117010. [PMID: 38941890 DOI: 10.1016/j.biopha.2024.117010] [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: 04/11/2024] [Revised: 05/29/2024] [Accepted: 06/17/2024] [Indexed: 06/30/2024] Open
Abstract
Diabetes mellitus (DM) is a metabolic disorder characterized by hyperglycemia, with its prevalence linked to both genetic predisposition and environmental factors. Epigenetic modifications, particularly through histone deacetylases (HDACs), have been recognized for their significant influence on DM pathogenesis. This review focuses on the classification of HDACs, their role in DM and its complications, and the potential therapeutic applications of HDAC inhibitors. HDACs, which modulate gene expression without altering DNA sequences, are categorized into four classes with distinct functions and tissue specificity. HDAC inhibitors (HDACi) have shown efficacy in various diseases, including DM, by targeting these enzymes. The review highlights how HDACs regulate β-cell function, insulin sensitivity, and hepatic gluconeogenesis in DM, as well as their impact on diabetic cardiomyopathy, nephropathy, and retinopathy. Finally, we suggest that targeted histone modification is expected to become a key method for the treatment of diabetes and its complications. The study of HDACi offers insights into new treatment strategies for DM and its associated complications.
Collapse
Affiliation(s)
- Li Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610072, PR China; Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, PR China
| | - Yuning Bai
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, PR China
| | - Zhengmin Cao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, PR China
| | - Ziwei Guo
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, PR China
| | - Yanjie Lian
- Department of Cardiovascular Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, PR China
| | - Pan Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610072, PR China
| | - Yixian Zeng
- Department of Proctology, Beibei Hospital of Traditional Chinese Medicine, Chongqing 400799, PR China
| | - Wenliang Lyu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, PR China.
| | - Qiu Chen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610072, PR China.
| |
Collapse
|
20
|
Chernysheva МB, Ruchko ЕS, Karimova МV, Vorotelyak ЕA, Vasiliev АV. Development, regeneration, and physiological expansion of functional β-cells: Cellular sources and regulators. Front Cell Dev Biol 2024; 12:1424278. [PMID: 39045459 PMCID: PMC11263198 DOI: 10.3389/fcell.2024.1424278] [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: 04/27/2024] [Accepted: 06/18/2024] [Indexed: 07/25/2024] Open
Abstract
Pancreatic regeneration is a complex process observed in both normal and pathological conditions. The aim of this review is to provide a comprehensive understanding of the emergence of a functionally active population of insulin-secreting β-cells in the adult pancreas. The renewal of β-cells is governed by a multifaceted interaction between cellular sources of genetic and epigenetic factors. Understanding the development and heterogeneity of β-cell populations is crucial for functional β-cell regeneration. The functional mass of pancreatic β-cells increases in situations such as pregnancy and obesity. However, the specific markers of mature β-cell populations and postnatal pancreatic progenitors capable of increasing self-reproduction in these conditions remain to be elucidated. The capacity to regenerate the β-cell population through various pathways, including the proliferation of pre-existing β-cells, β-cell neogenesis, differentiation of β-cells from a population of progenitor cells, and transdifferentiation of non-β-cells into β-cells, reveals crucial molecular mechanisms for identifying cellular sources and inducers of functional cell renewal. This provides an opportunity to identify specific cellular sources and mechanisms of regeneration, which could have clinical applications in treating various pathologies, including in vitro cell-based technologies, and deepen our understanding of regeneration in different physiological conditions.
Collapse
Affiliation(s)
- М. B. Chernysheva
- Cell Biology Laboratory, Koltzov Institute of Developmental Biology, Moscow, Russia
| | - Е. S. Ruchko
- Cell Biology Laboratory, Koltzov Institute of Developmental Biology, Moscow, Russia
| | - М. V. Karimova
- Cell Biology Laboratory, Koltzov Institute of Developmental Biology, Moscow, Russia
- Department of Biology and Biotechnologies Charles Darwin, The Sapienza University of Rome, Rome, Italy
| | - Е. A. Vorotelyak
- Cell Biology Laboratory, Koltzov Institute of Developmental Biology, Moscow, Russia
| | - А. V. Vasiliev
- Cell Biology Laboratory, Koltzov Institute of Developmental Biology, Moscow, Russia
| |
Collapse
|
21
|
Ampofo E, Pack M, Wrublewsky S, Boewe AS, Spigelman AF, Koch H, MacDonald PE, Laschke MW, Montenarh M, Götz C. CK2 activity is crucial for proper glucagon expression. Diabetologia 2024; 67:1368-1385. [PMID: 38503901 PMCID: PMC11153270 DOI: 10.1007/s00125-024-06128-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 02/07/2024] [Indexed: 03/21/2024]
Abstract
AIMS/HYPOTHESIS Protein kinase CK2 acts as a negative regulator of insulin expression in pancreatic beta cells. This action is mainly mediated by phosphorylation of the transcription factor pancreatic and duodenal homeobox protein 1 (PDX1). In pancreatic alpha cells, PDX1 acts in a reciprocal fashion on glucagon (GCG) expression. Therefore, we hypothesised that CK2 might positively regulate GCG expression in pancreatic alpha cells. METHODS We suppressed CK2 kinase activity in αTC1 cells by two pharmacological inhibitors and by the CRISPR/Cas9 technique. Subsequently, we analysed GCG expression and secretion by real-time quantitative RT-PCR, western blot, luciferase assay, ELISA and DNA pull-down assays. We additionally studied paracrine effects on GCG secretion in pseudoislets, isolated murine islets and human islets. In vivo, we examined the effect of CK2 inhibition on blood glucose levels by systemic and alpha cell-specific CK2 inhibition. RESULTS We found that CK2 downregulation reduces GCG secretion in the murine alpha cell line αTC1 (e.g. from 1094±124 ng/l to 459±110 ng/l) by the use of the CK2-inhibitor SGC-CK2-1. This was due to a marked decrease in Gcg gene expression through alteration of the binding of paired box protein 6 (PAX6) and transcription factor MafB to the Gcg promoter. The analysis of the underlying mechanisms revealed that both transcription factors are displaced by PDX1. Ex vivo experiments in isolated murine islets and pseudoislets further demonstrated that CK2-mediated reduction in GCG secretion was only slightly affected by the higher insulin secretion after CK2 inhibition. The kidney capsule transplantation model showed the significance of CK2 for GCG expression and secretion in vivo. Finally, CK2 downregulation also reduced the GCG secretion in islets isolated from humans. CONCLUSIONS/INTERPRETATION These novel findings not only indicate an important function of protein kinase CK2 for proper GCG expression but also demonstrate that CK2 may be a promising target for the development of novel glucose-lowering drugs.
Collapse
Affiliation(s)
- Emmanuel Ampofo
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Mandy Pack
- Medical Biochemistry and Molecular Biology, Saarland University, Homburg, Germany
| | - Selina Wrublewsky
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Anne S Boewe
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Aliya F Spigelman
- Department of Pharmacology and Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Hanna Koch
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Patrick E MacDonald
- Department of Pharmacology and Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Matthias W Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Mathias Montenarh
- Medical Biochemistry and Molecular Biology, Saarland University, Homburg, Germany
| | - Claudia Götz
- Medical Biochemistry and Molecular Biology, Saarland University, Homburg, Germany.
| |
Collapse
|
22
|
Beyer ASL, Kaemmerer D, Sänger J, Lupp A. Expression of FAM159B in Humans, Rats, and Mice: A Cross-species Examination. J Histochem Cytochem 2024; 72:467-487. [PMID: 38907656 PMCID: PMC11308193 DOI: 10.1369/00221554241262368] [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] [Accepted: 05/23/2024] [Indexed: 06/24/2024] Open
Abstract
Little is known about the adaptor protein FAM159B. To determine whether FAM159B expression findings in rats or mice can be extrapolated to humans, we compared FAM159B expression in healthy tissue samples from all three species using immunohistochemistry. Despite variations in expression intensity, similar FAM159B expression patterns were observed in most organs across species. The most prominent species difference was noted in pancreatic islets; while FAM159B expression was limited to single cells on the outer edges in mice and rats, it was detectable across entire islets in humans. Double-labeling immunohistochemistry revealed partial overlap of FAM159B expression with that of insulin, glucagon, and somatostatin in human islets. By contrast, FAM159B showed complete colocalization with only somatostatin in rats and mice. An additional analysis of FAM159B expression in lean and obese Zucker rats revealed larger islet areas due to increased β-cell mass in obese rats, which was accompanied by a smaller percentage of FAM159B-positive δ-cells per islet area. Beyond the known differences in islet architecture across species, our results point to larger dissimilarities in blood glucose regulation between rodents and humans than generally assumed. Moreover, findings regarding FAM159B expression (and function) cannot be directly transferred between rodents and humans.
Collapse
Affiliation(s)
| | - Daniel Kaemmerer
- Department of General and Visceral Surgery, Zentralklinik Bad Berka, Bad Berka, Germany
| | - Jörg Sänger
- Laboratory of Pathology and Cytology Bad Berka, Bad Berka, Germany
| | - Amelie Lupp
- Institute of Pharmacology and Toxicology, Jena University Hospital, Jena, Germany
| |
Collapse
|
23
|
Fuyuki A, Sohel MSH, Homma T, Kitamura K, Takashima S, Onouchi S, Saito S. Selective prosaposin expression in Langerhans islets of the mouse pancreas. Tissue Cell 2024; 88:102367. [PMID: 38537378 DOI: 10.1016/j.tice.2024.102367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 03/12/2024] [Accepted: 03/22/2024] [Indexed: 06/17/2024]
Abstract
The islets of Langerhans are clusters of endocrine cells surrounded by exocrine acinar cells in the pancreas. Prosaposin is a housekeeping protein required for normal lysosomal function, but its expression level is significantly different among tissues. Prosaposin also exists in various body fluids including serum. Intracellularly, prosaposin activates lysosomes and may support autophagy, and extracellularly, prosaposin promotes survival of neurons via G protein-coupled receptors. In this study, prosaposin and its mRNA expression were examined in endocrine cells of the islets as well as in exocrine acinar cells in the pancreas of mice by in situ hybridization and immunostaining. High expression levels of prosaposin were found in Alpha, Beta and Delta cells in the islets, whereas prosaposin mRNA expression was faint or negative and prosaposin immunoreactivity was negative in exocrine acinar cells. The high expression levels of prosaposin in endocrine cells may indicate that prosaposin plays a crucial role in crinophagy, which is a characteristic autophagy in peptide-secreting endocrine cells, and/or that prosaposin is secreted from pancreatic islets. Since prosaposin has been reported in serum, this study suggests a new possible function of the Langerhans islets.
Collapse
Affiliation(s)
- Aimi Fuyuki
- Laboratory of Veterinary Anatomy, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan; Laboratory of Veterinary Anatomy, Joint Graduate School of Veterinary Science, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Md Shahriar Hasan Sohel
- Laboratory of Veterinary Anatomy, Joint Graduate School of Veterinary Science, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Takeshi Homma
- Laboratory of Veterinary Anatomy, Joint Graduate School of Veterinary Science, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Kai Kitamura
- Laboratory of Veterinary Anatomy, Joint Graduate School of Veterinary Science, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Shigeo Takashima
- Division of Genomics Research, Life Science Research Center, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Sawa Onouchi
- Laboratory of Veterinary Anatomy, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan; Laboratory of Veterinary Anatomy, Joint Graduate School of Veterinary Science, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Shouichiro Saito
- Laboratory of Veterinary Anatomy, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan; Laboratory of Veterinary Anatomy, Joint Graduate School of Veterinary Science, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan.
| |
Collapse
|
24
|
Tansi FL, Schrepper A, Schwarzer M, Teichgräber U, Hilger I. Identifying the Morphological and Molecular Features of a Cell-Based Orthotopic Pancreatic Cancer Mouse Model during Growth over Time. Int J Mol Sci 2024; 25:5619. [PMID: 38891809 PMCID: PMC11171605 DOI: 10.3390/ijms25115619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 06/21/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), characterized by hypovascularity, hypoxia, and desmoplastic stroma is one of the deadliest malignancies in humans, with a 5-year survival rate of only 7%. The anatomical location of the pancreas and lack of symptoms in patients with early onset of disease accounts for late diagnosis. Consequently, 85% of patients present with non-resectable, locally advanced, or advanced metastatic disease at diagnosis and rely on alternative therapies such as chemotherapy, immunotherapy, and others. The response to these therapies highly depends on the stage of disease at the start of therapy. It is, therefore, vital to consider the stages of PDAC models in preclinical studies when testing new therapeutics and treatment modalities. We report a standardized induction of cell-based orthotopic pancreatic cancer models in mice and the identification of vital features of their progression by ultrasound imaging and histological analysis of the level of pancreatic stellate cells, mature fibroblasts, and collagen. The results highlight that early-stage primary tumors are secluded in the pancreas and advance towards infiltrating the omentum at week 5-7 post implantation of the BxPC-3 and Panc-1 models investigated. Late stages show extensive growth, the infiltration of the omentum and/or stomach wall, metastases, augmented fibroblasts, and collagen levels. The findings can serve as suggestions for defining growth parameter-based stages of orthotopic pancreatic cancer models for the preclinical testing of drug efficacy in the future.
Collapse
Affiliation(s)
- Felista L. Tansi
- Experimental Radiology, Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Andrea Schrepper
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany (M.S.)
| | - Michael Schwarzer
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany (M.S.)
| | - Ulf Teichgräber
- Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Ingrid Hilger
- Experimental Radiology, Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany
| |
Collapse
|
25
|
Li Y, Gao J, Li Y, Duan X, Shen C. Non-specific uptake of 18F-FAPI-04 in the pancreas and its related factors: a post-hoc analysis of an ongoing prospective clinical trial. Sci Rep 2024; 14:11141. [PMID: 38750103 PMCID: PMC11096165 DOI: 10.1038/s41598-024-62005-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 05/13/2024] [Indexed: 05/18/2024] Open
Abstract
This study aimed to analyze the characteristics of the non-specific uptake (NSU) of 18F-labeled fibroblast activation protein inhibitor (18F-FAPI) of the pancreas and investigate the related factors. Totally, 78 patients who underwent both 18F-fluorodeoxyglucose (FDG) and 18F-FAPI PET/CT examinations were divided into normal (n = 53) and NSU (n = 25) groups. The differences in general information, medical history, laboratory indexes and uptake were compared. Receiver operating characteristic (ROC) curves were used to analyze the optimal cut-off values. The correlations between 18F-FAPI-SUVmax and blood cell analysis, liver function indexes, tumor markers, and inflammatory indices were analyzed. The logistic regression model was used to estimate the independent factors. Both 18F-FAPI (4.48 ± 0.98 vs. 2.01 ± 0.53, t = 11.718, P < 0.05) and 18F-FDG (2.23 ± 0.42 vs. 2.02 ± 0.44, t = 2.036, P = 0.045) showed significantly higher in NSU group. Patients in the NSU group tended to be complicated with a history of drinking (P = 0.034), chronic liver diseases (P = 0.006), and surgery of gastrectomy (P = 0.004). ROC analysis showed cutoff values of 3.25 and 2.05 for 18F-FAPI and 18F-FDG in identifying the NSU. Patients in the NSU group showed less platelet count, higher platelet volume, higher total bilirubin, direct or indirect bilirubin (P < 0.05). Platelet count, platelet crit, large platelet ratio, aspartate aminotransferase (AST), α-L-fucosidase, and total, direct or indirect bilirubin were correlated with 18F-FAPI-SUVmax (P < 0.05). AST [1.099 (1.014, 1.192), P = 0.021] and total bilirubin [1.137 (1.035, 1.249), P = 0.007] were two independent factors in the step forward logistic regression, and platelet/% [1.079 (1.004, 1.160), P = 0.039] and total bilirubin [1.459 (1.016, 2.095), P = 0.041] were two independent factors in the step backward logistic regression for the prediction of pancreatic uptake of 18F-FAPI. 18F-FAPI-PET/CT was better than 18F-FDG in predicting the pancreatic NSU, and NSU is related to a history of drinking, chronic liver diseases, gastrectomy, heteromorphic platelet, and impaired liver function.
Collapse
Affiliation(s)
- Yan Li
- Department of PET/CT, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Jungang Gao
- Department of PET/CT, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Yang Li
- Department of PET/CT, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Xiaoyi Duan
- Department of PET/CT, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Cong Shen
- Department of PET/CT, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
26
|
Tanner AR, Kennedy VC, Lynch CS, Winger QA, Anthony RV, Rozance PJ. Increasing maternal glucose concentrations is insufficient to restore placental glucose transfer in chorionic somatomammotropin RNA interference pregnancies. Am J Physiol Endocrinol Metab 2024; 326:E602-E615. [PMID: 38353640 PMCID: PMC11376830 DOI: 10.1152/ajpendo.00331.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 02/01/2024] [Accepted: 02/05/2024] [Indexed: 04/19/2024]
Abstract
We previously demonstrated impaired placental nutrient transfer in chorionic somatomammotropin (CSH) RNA interference (RNAi) pregnancies, with glucose transfer being the most impacted. Thus, we hypothesized that despite experimentally elevating maternal glucose, diminished umbilical glucose uptake would persist in CSH RNAi pregnancies, demonstrating the necessity of CSH for adequate placental glucose transfer. Trophectoderm of sheep blastocysts (9 days of gestational age; dGA) were infected with a lentivirus expressing either nontargeting control (CON RNAi; n = 5) or CSH-specific shRNA (CSH RNAi; n = 7) before transfer into recipient sheep. At 126 dGA, pregnancies were fitted with vascular catheters and underwent steady-state metabolic studies (3H2O transplacental diffusion) at 137 ± 0 dGA, before and during a maternal hyperglycemic clamp. Umbilical glucose and oxygen uptakes, as well as insulin and IGF1 concentrations, were impaired (P ≤ 0.01) in CSH RNAi fetuses and were not rescued by elevated maternal glucose. This is partially due to impaired uterine and umbilical blood flow (P ≤ 0.01). However, uteroplacental oxygen utilization was greater (P ≤ 0.05) during the maternal hyperglycemic clamp, consistent with greater placental oxidation of substrates. The relationship between umbilical glucose uptake and the maternal-fetal glucose gradient was analyzed, and while the slope (CON RNAi, Y = 29.54X +74.15; CSH RNAi, Y = 19.05X + 52.40) was not different, the y-intercepts and elevation were (P = 0.003), indicating reduced maximal glucose transport during maternal hyperglycemia. Together, these data suggested that CSH plays a key role in modulating placental metabolism that ultimately promotes maximal placental glucose transfer.NEW & NOTEWORTHY The current study demonstrated a novel, critical autocrine role for chorionic somatomammotropin in augmenting placental glucose transfer and maintaining placental oxidative metabolism. In pregnancies with CSH deficiency, excess glucose in maternal circulation is insufficient to overcome fetal hypoglycemia due to impaired placental glucose transfer and elevated placental metabolic demands. This suggests that perturbations in glucose transfer in CSH RNAi pregnancies are due to compromised metabolic efficiency along with reduced placental mass.
Collapse
Affiliation(s)
- Amelia R Tanner
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States
- Perinatal Research Center, University of Colorado School of Medicine, Aurora, Colorado, United States
| | - Victoria C Kennedy
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States
| | - Cameron S Lynch
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States
| | - Quinton A Winger
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States
| | - Russell V Anthony
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States
| | - Paul J Rozance
- Perinatal Research Center, University of Colorado School of Medicine, Aurora, Colorado, United States
| |
Collapse
|
27
|
Fernandes‐da‐Silva A, Miranda RA, Lisboa PC, Souza‐Mello V. Revisiting pancreatic islet isolation in murine models: A practical and effective technical protocol. Physiol Rep 2024; 12:e16040. [PMID: 38725080 PMCID: PMC11082087 DOI: 10.14814/phy2.16040] [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: 02/15/2024] [Revised: 04/20/2024] [Accepted: 04/20/2024] [Indexed: 05/12/2024] Open
Abstract
The endocrine pancreas is composed of clusters of cell groups called pancreatic islets. These cells are responsible for the synthesis and secretion of hormones crucial for glycemic homeostasis, such as insulin and glucagon. Therefore, these cells were the targets of many studies. One method to study and/or understand endocrine pancreatic physiology is the isolation of these islets and stimulation of hormone production using different concentrations of glucose, agonists, and/or antagonists of specific secretagogues and mimicking the stimulation of hormonal synthesis and secretion. Many researchers studied pancreatic physiology in murine models due to their ease of maintenance and rapid development. However, the isolation of pancreatic islets involves meticulous processes that may vary between rodent species. The present study describes a simple and effective technical protocol for isolating intact islets from mice and rats for use as a practical guide for researchers. The method involves digestion of the acinar parenchyma by intraductal collagenase. Isolated islets are suitable for in vitro endocrine secretion analyses, microscopy techniques, and biochemical analyses.
Collapse
Affiliation(s)
- Aline Fernandes‐da‐Silva
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology Roberto Alcantara GomesState University of Rio de JaneiroRio de JaneiroBrazil
| | - Rosiane Aparecida Miranda
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Institute of Biology Roberto Alcantara GomesState University of Rio de JaneiroRio de JaneiroBrazil
| | - Patricia Cristina Lisboa
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Institute of Biology Roberto Alcantara GomesState University of Rio de JaneiroRio de JaneiroBrazil
| | - Vanessa Souza‐Mello
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology Roberto Alcantara GomesState University of Rio de JaneiroRio de JaneiroBrazil
| |
Collapse
|
28
|
Holmberg SR, Sakamoto Y, Kato A, Romero MF. The role of Na +-coupled bicarbonate transporters (NCBT) in health and disease. Pflugers Arch 2024; 476:479-503. [PMID: 38536494 PMCID: PMC11338471 DOI: 10.1007/s00424-024-02937-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 04/11/2024]
Abstract
Cellular and organism survival depends upon the regulation of pH, which is regulated by highly specialized cell membrane transporters, the solute carriers (SLC) (For a comprehensive list of the solute carrier family members, see: https://www.bioparadigms.org/slc/ ). The SLC4 family of bicarbonate (HCO3-) transporters consists of ten members, sorted by their coupling to either sodium (NBCe1, NBCe2, NBCn1, NBCn2, NDCBE), chloride (AE1, AE2, AE3), or borate (BTR1). The ionic coupling of SLC4A9 (AE4) remains controversial. These SLC4 bicarbonate transporters may be controlled by cellular ionic gradients, cellular membrane voltage, and signaling molecules to maintain critical cellular and systemic pH (acid-base) balance. There are profound consequences when blood pH deviates even a small amount outside the normal range (7.35-7.45). Chiefly, Na+-coupled bicarbonate transporters (NCBT) control intracellular pH in nearly every living cell, maintaining the biological pH required for life. Additionally, NCBTs have important roles to regulate cell volume and maintain salt balance as well as absorption and secretion of acid-base equivalents. Due to their varied tissue expression, NCBTs have roles in pathophysiology, which become apparent in physiologic responses when their expression is reduced or genetically deleted. Variations in physiological pH are seen in a wide variety of conditions, from canonically acid-base related conditions to pathologies not necessarily associated with acid-base dysfunction such as cancer, glaucoma, or various neurological diseases. The membranous location of the SLC4 transporters as well as recent advances in discovering their structural biology makes them accessible and attractive as a druggable target in a disease context. The role of sodium-coupled bicarbonate transporters in such a large array of conditions illustrates the potential of treating a wide range of disease states by modifying function of these transporters, whether that be through inhibition or enhancement.
Collapse
Affiliation(s)
- Shannon R Holmberg
- Physiology & Biomedical Engineering, Mayo Clinic College of Medicine & Science, 200 1st Street SW, Rochester, MN 55905, USA
- Biochemistry & Molecular Biology, Mayo Clinic College of Medicine & Science, 200 1st Street SW, Rochester, MN, USA
| | - Yohei Sakamoto
- School of Life Science and Technology, Tokyo Institute of Technology, Midori-Ku, Yokohama, 226-8501, Japan
| | - Akira Kato
- School of Life Science and Technology, Tokyo Institute of Technology, Midori-Ku, Yokohama, 226-8501, Japan
| | - Michael F Romero
- Physiology & Biomedical Engineering, Mayo Clinic College of Medicine & Science, 200 1st Street SW, Rochester, MN 55905, USA.
- Nephrology & Hypertension, Mayo Clinic College of Medicine & Science, 200 1st Street SW, Rochester, MN, USA.
| |
Collapse
|
29
|
Villaca CBP, Mastracci TL. Pancreatic Crosstalk in the Disease Setting: Understanding the Impact of Exocrine Disease on Endocrine Function. Compr Physiol 2024; 14:5371-5387. [PMID: 39109973 PMCID: PMC11425433 DOI: 10.1002/cphy.c230008] [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] [Indexed: 08/15/2024]
Abstract
The exocrine and endocrine are functionally distinct compartments of the pancreas that have traditionally been studied as separate entities. However, studies of embryonic development, adult physiology, and disease pathogenesis suggest there may be critical communication between exocrine and endocrine cells. In fact, the incidence of the endocrine disease diabetes secondary to exocrine disease/dysfunction ranges from 25% to 80%, depending on the type and severity of the exocrine pathology. Therefore, it is necessary to investigate how exocrine-endocrine "crosstalk" may impact pancreatic function. In this article, we discuss common exocrine diseases, including cystic fibrosis, acute, hereditary, and chronic pancreatitis, and the impact of these exocrine diseases on endocrine function. Additionally, we review how obesity and fatty pancreas influence exocrine function and the impact on cellular communication between the exocrine and endocrine compartments. Interestingly, in all pathologies, there is evidence that signals from the exocrine disease contribute to endocrine dysfunction and the progression to diabetes. Continued research efforts to identify the mechanisms that underlie the crosstalk between various cell types in the pancreas are critical to understanding normal pancreatic physiology as well as disease states. © 2024 American Physiological Society. Compr Physiol 14:5371-5387, 2024.
Collapse
Affiliation(s)
| | - Teresa L Mastracci
- Department of Biology, Indiana University Indianapolis, Indianapolis, Indiana, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
30
|
Lipovšek S, Dolenšek J, Dariš B, Valladolid-Acebes I, Vajs T, Leitinger G, Stožer A, Skelin Klemen M. Western diet-induced ultrastructural changes in mouse pancreatic acinar cells. Front Cell Dev Biol 2024; 12:1380564. [PMID: 38550379 PMCID: PMC10972872 DOI: 10.3389/fcell.2024.1380564] [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: 02/01/2024] [Accepted: 03/05/2024] [Indexed: 12/18/2024] Open
Abstract
Mouse models of diet-induced type 2 diabetes mellitus provide powerful tools for studying the structural and physiological changes that are related to the disease progression. In this study, diabetic-like glucose dysregulation was induced in mice by feeding them a western diet, and light and transmission electron microscopy were used to study the ultrastructural changes in the pancreatic acinar cells. Acinar necrosis and vacuolization of the cytoplasm were the most prominent features. Furthermore, we observed intracellular and extracellular accumulation of lipid compounds in the form of lipid droplets, structural enlargement of the cisternae of the rough endoplasmic reticulum (RER), and altered mitochondrial morphology, with mitochondria lacking the typical organization of the inner membrane. Last, autophagic structures, i.e., autophagosomes, autolysosomes, and residual bodies, were abundant within the acinar cells of western diet-fed mice, and the autolysosomes contained lipids and material of varying electron density. While diets inducing obesity and type 2 diabetes are clearly associated with structural changes and dysfunction of the endocrine pancreas, we here demonstrate the strong effect of dietary intervention on the structure of acinar cells in the exocrine part of the organ before detectable changes in plasma amylase activity, which may help us better understand the development of non-alcoholic fatty pancreas disease and its association with endo- and exocrine dysfunction.
Collapse
Affiliation(s)
- Saška Lipovšek
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Department of Biology, Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
- Gottfried Schatz Research Center, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
- Faculty of Chemistry and Chemical Engineering, University of Maribor, Maribor, Slovenia
| | - Jurij Dolenšek
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Department of Biology, Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
| | - Barbara Dariš
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Ismael Valladolid-Acebes
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Tanja Vajs
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Gerd Leitinger
- Gottfried Schatz Research Center, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Andraž Stožer
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | | |
Collapse
|
31
|
Mohanan A, Biju P, V B, V G. Unraveling Proto-Oncogene (ErbB2) Expression in Patients With Carcinoma Head of Pancreas and Chronic Pancreatitis Patients: A Case-Control Study. Cureus 2024; 16:e54859. [PMID: 38533139 PMCID: PMC10964396 DOI: 10.7759/cureus.54859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2024] [Indexed: 03/28/2024] Open
Abstract
Background The pre-malignant tendency of the normal, non-affected portion of the pancreas is not as well explored as the multicentricity documented in pancreatic cancer cases. In order to ascertain the expression of inflammatory markers and Erythroblastic Oncogene B (ErbB2) in the non-affected pancreas in patients with pancreatic cancer, a case-control study was carried out. Materials and methods In patients who underwent pancreatoduodenectomy for pancreatic cancer (PC), pro-inflammatory genes and a tumor marker, erythroblastic oncogene 2 (ErbB2) in the epidermal growth factor receptor family were analyzed in the pancreatic tissue at the cut surface of the normal pancreas using qRT-PCR. Twenty patients diagnosed with Chronic pancreatitis (CP) after Frey's surgical procedure were selected, and their pancreatic tissues were analyzed as controls. The HPLC-purified primers were designed using National Center for Biotechnology Information (NCBI) software. The primer's specificity was verified for gene expression analysis using the Basic Local Alignment Search Tool (BLAST). The genes under study were normalized using β-actin as the housekeeping gene, and the 2-ddct method was used to compute the fold change compared to the control sample. Results Patients with margin-positive were not included. Pro-inflammatory genes (TNF-α, NF-kβ, and COX-2) had significantly lower foldchange in PC patients compared to the CP group. The CP control group had higher levels of IL-6 gene expression than the PC group. Patients with pancreatic cancer had a considerably higher expression of the ErbB2 gene than patients with CP. Conclusion The upregulated ErbB2 gene in the unaffected pancreatic tissue of pancreatic cancer patients, when compared to controls, indicates that the remaining pancreas may have the capacity to cause cancer. Proto-oncogene may play a role in the pathophysiologic process in patients with pancreatic cancer.
Collapse
Affiliation(s)
- Abhina Mohanan
- Surgical Gastroenterology, Jawaharlal Institute of Postgraduate Medical Education & Research, Puducherry, IND
| | - Pottakkat Biju
- Surgical Gastroenterology, Jawaharlal Institute of Postgraduate Medical Education & Research, Puducherry, IND
| | - Balasubramaniyan V
- Biochemistry, Jawaharlal Institute of Postgraduate Medical Education & Research, Puducherry, IND
| | - Gladwin V
- Anatomy, Jawaharlal Institute of Postgraduate Medical Education & Research, Puducherry, IND
| |
Collapse
|
32
|
Sallam NG, Boraie NA, Sheta E, El-Habashy SE. Targeted delivery of genistein for pancreatic cancer treatment using hyaluronic-coated cubosomes bioactivated with frankincense oil. Int J Pharm 2024; 649:123637. [PMID: 38008234 DOI: 10.1016/j.ijpharm.2023.123637] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 11/01/2023] [Accepted: 11/22/2023] [Indexed: 11/28/2023]
Abstract
Pancreatic cancer is an aggressive malignancy that remains a major cause of cancer-related deaths. Research for innovative anticancer therapeutic options is thus imperative. In this regard, phytotherapeutics offer great promise as efficient treatment modalities, especially leveraging nanodrug delivery. Herein, we innovatively coloaded the flavonoid genistein (Gen) and frankincense essential oil (FO) within cubosomes, which were then coated with the bioactive ligand hyaluronic acid (HA/Gen-FO-Cub) for active-targeting of pancreatic cancer. The novel HA/Gen-FO-Cub displayed optimum nanosize (198.2 ± 4.5 nm), PDI (0.27 ± 0.01), zeta-potential (-34.7 ± 1.2 mV), Gen entrapment (99.3 ± 0.01 %), and controlled Gen release (43.7 ± 1.2 % after 120 h). HA/Gen-FO-Cub exerted selective anticancer activity on pancreatic cancer cells (PANC-1; 8-fold drop in IC50), cellular uptake and anti-migratory effect compared to Gen solution. HA/Gen-FO-Cub revealed prominent cytocompatibility (100 ± 5.9 % viability of human dermal fibroblast). Moreover, HA/Gen-FO-Cub boosted the in vivo anticancer activity of Gen in an orthotopic cancer model, affording tumor growth suppression (2.5-fold drop) and downregulation of NFκB and VEGF (2.9- and 1.8-fold decrease, respectively), compared to Gen suspension. Antimetastatic efficacy and Bcl-2-downexpression was histologically confirmed. Our findings demonstrate the promising anticancer aptitude of HA/Gen-FO-Cub as an effective phytotherapeutic nanodelivery system for pancreatic cancer therapy.
Collapse
Affiliation(s)
- Nourhan G Sallam
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Nabila A Boraie
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Eman Sheta
- Pathology Department, Faculty of Medicine, Alexandria University, Alexandria 21131, Egypt
| | - Salma E El-Habashy
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt.
| |
Collapse
|
33
|
Coppola A, Grasso D, Fontana F, Piacentino F, Minici R, Laganà D, Ierardi AM, Carrafiello G, D’Angelo F, Carcano G, Venturini M. Innovative Experimental Ultrasound and US-Related Techniques Using the Murine Model in Pancreatic Ductal Adenocarcinoma: A Systematic Review. J Clin Med 2023; 12:7677. [PMID: 38137745 PMCID: PMC10743777 DOI: 10.3390/jcm12247677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/24/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a cancer with one of the highest mortality rates in the world. Several studies have been conductedusing preclinical experiments in mice to find new therapeutic strategies. Experimental ultrasound, in expert hands, is a safe, multifaceted, and relatively not-expensive device that helps researchers in several ways. In this systematic review, we propose a summary of the applications of ultrasonography in a preclinical mouse model of PDAC. Eighty-eight studies met our inclusion criteria. The included studies could be divided into seven main topics: ultrasound in pancreatic cancer diagnosis and progression (n: 21); dynamic contrast-enhanced ultrasound (DCE-US) (n: 5); microbubble ultra-sound-mediated drug delivery; focused ultrasound (n: 23); sonodynamic therapy (SDT) (n: 7); harmonic motion elastography (HME) and shear wave elastography (SWE) (n: 6); ultrasound-guided procedures (n: 9). In six cases, the articles fit into two or more sections. In conclusion, ultrasound can be a really useful, eclectic, and ductile tool in different diagnostic areas, not only regarding diagnosis but also in therapy, pharmacological and interventional treatment, and follow-up. All these multiple possibilities of use certainly represent a good starting point for the effective and wide use of murine ultrasonography in the study and comprehensive evaluation of pancreatic cancer.
Collapse
Affiliation(s)
- Andrea Coppola
- Diagnostic and Interventional Radiology Unit, Circolo Hospital, ASST Sette Laghi, 21100 Varese, Italy (M.V.)
- Department of Medicine and Technological Innovation, Insubria University, 21100 Varese, Italy
| | - Dario Grasso
- Diagnostic and Interventional Radiology Unit, Circolo Hospital, ASST Sette Laghi, 21100 Varese, Italy (M.V.)
- Department of Medicine and Technological Innovation, Insubria University, 21100 Varese, Italy
| | - Federico Fontana
- Diagnostic and Interventional Radiology Unit, Circolo Hospital, ASST Sette Laghi, 21100 Varese, Italy (M.V.)
- Department of Medicine and Technological Innovation, Insubria University, 21100 Varese, Italy
| | - Filippo Piacentino
- Diagnostic and Interventional Radiology Unit, Circolo Hospital, ASST Sette Laghi, 21100 Varese, Italy (M.V.)
- Department of Medicine and Technological Innovation, Insubria University, 21100 Varese, Italy
| | - Roberto Minici
- Radiology Unit, Dulbecco University Hospital, 88100 Catanzaro, Italy; (R.M.)
| | - Domenico Laganà
- Radiology Unit, Dulbecco University Hospital, 88100 Catanzaro, Italy; (R.M.)
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy
| | - Anna Maria Ierardi
- Radiology Unit, IRCCS Ca Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | | | - Fabio D’Angelo
- Department of Medicine and Surgery, Insubria University, 21100 Varese, Italy;
- Orthopedic Surgery Unit, ASST Sette Laghi, 21100 Varese, Italy
| | - Giulio Carcano
- Department of Medicine and Technological Innovation, Insubria University, 21100 Varese, Italy
- Emergency and Transplant Surgery Department, ASST Sette Laghi, 21100 Varese, Italy
| | - Massimo Venturini
- Diagnostic and Interventional Radiology Unit, Circolo Hospital, ASST Sette Laghi, 21100 Varese, Italy (M.V.)
- Department of Medicine and Technological Innovation, Insubria University, 21100 Varese, Italy
| |
Collapse
|
34
|
Li Y, Huang J, Chen Y, Zhu S, Huang Z, Yang L, Li G. Nerve function restoration following targeted muscle reinnervation after varying delayed periods. Neural Regen Res 2023; 18:2762-2766. [PMID: 37449642 DOI: 10.4103/1673-5374.373659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023] Open
Abstract
Targeted muscle reinnervation has been proposed for reconstruction of neuromuscular function in amputees. However, it is unknown whether performing delayed targeted muscle reinnervation after nerve injury will affect restoration of function. In this rat nerve injury study, the median and musculocutaneous nerves of the forelimb were transected. The proximal median nerve stump was sutured to the distal musculocutaneous nerve stump immediately and 2 and 4 weeks after surgery to reinnervate the biceps brachii. After targeted muscle reinnervation, intramuscular myoelectric signals from the biceps brachii were recorded. Signal amplitude gradually increased with time. Biceps brachii myoelectric signals and muscle fiber morphology and grooming behavior did not significantly differ among rats subjected to delayed target muscle innervation for different periods. Targeted muscle reinnervation delayed for 4 weeks can acquire the same nerve function restoration effect as that of immediate reinnervation.
Collapse
Affiliation(s)
- Yuanheng Li
- Key Laboratory of Human-Machine Intelligence-Synergy Systems and Branch of Shenzhen Institute of Artificial Intelligence and Robotics for Society, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
| | - Jiangping Huang
- Key Laboratory of Human-Machine Intelligence-Synergy Systems and Branch of Shenzhen Institute of Artificial Intelligence and Robotics for Society, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
| | - Yuling Chen
- Department of Rehabilitation Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province; Department of Rehabilitation Medicine, Yibin Hospital of Traditional Chinese Medicine, Yibin, Sichuan Province, China
| | - Shanshan Zhu
- Key Laboratory of Human-Machine Intelligence-Synergy Systems and Branch of Shenzhen Institute of Artificial Intelligence and Robotics for Society, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
| | - Zhen Huang
- Department of Rehabilitation Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province; Department of Rehabilitation Medicine, Yibin Hospital of Traditional Chinese Medicine, Yibin, Sichuan Province, China
| | - Lin Yang
- Key Laboratory of Human-Machine Intelligence-Synergy Systems and Branch of Shenzhen Institute of Artificial Intelligence and Robotics for Society, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
| | - Guanglin Li
- Key Laboratory of Human-Machine Intelligence-Synergy Systems and Branch of Shenzhen Institute of Artificial Intelligence and Robotics for Society, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
| |
Collapse
|
35
|
Damiana TST, Paraïso P, de Ridder C, Stuurman D, Seimbille Y, Dalm SU. Side-by-side comparison of the two widely studied GRPR radiotracers, radiolabeled NeoB and RM2, in a preclinical setting. Eur J Nucl Med Mol Imaging 2023; 50:3851-3861. [PMID: 37584725 PMCID: PMC10611828 DOI: 10.1007/s00259-023-06364-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/24/2023] [Indexed: 08/17/2023]
Abstract
INTRODUCTION NeoB and RM2 are the most investigated gastrin-releasing peptide receptor (GRPR)-targeting radiotracers in preclinical and clinical studies. Therefore, an extensive side-by-side comparison of the two radiotracers is valuable to demonstrate whether one has advantages over the other. Accordingly, this study aims to compare the in vitro and in vivo characteristics of radiolabeled NeoB and RM2 to guide future clinical studies. METHOD The stability of the radiolabeled GRPR analogs was determined in phosphate buffered saline (PBS), and commercially available mouse and human serum. Target affinity was determined by incubating human prostate cancer PC-3 cells with [177Lu]Lu-NeoB or [177Lu]Lu-RM2, + / - increasing concentrations of unlabeled NeoB, RM2, or Tyr4-bombesin (BBN). To determine uptake and specificity cells were incubated with [177Lu]Lu-NeoB or [177Lu]Lu-RM2 + / - Tyr4-BBN. Moreover, in vivo studies were performed to determine biodistribution and pharmacokinetics. Finally, radiotracer binding to various GRPR-expressing human cancer tissues was investigated. RESULTS Both radiotracers demonstrated high stability in PBS and human serum, but stability in mouse serum decreased substantially over time. Moreover, both radiotracers demonstrated high GRPR affinity and specificity, but a higher uptake of [177Lu]Lu-NeoB was observed in in vitro studies. In vivo, no difference in tumor uptake was seen. The most prominent difference in uptake in physiological organs was observed in the GRPR-expressing pancreas; [177Lu]Lu-RM2 had less pancreatic uptake and a shorter pancreatic half-life than [177Lu]Lu-NeoB. Furthermore, [177Lu]Lu-RM2 presented with a lower tumor-to-kidney ratio, while the tumor-to-blood ratio was lower for [177Lu]Lu-NeoB. The autoradiography studies revealed higher binding of radiolabeled NeoB to all human tumor tissues. CONCLUSION Based on these findings, we conclude that the in vivo tumor-targeting capability of radiolabeled NeoB and RM2 is similar. Additional studies are needed to determine whether the differences observed in physiological organ uptakes, i.e., the pancreas, kidneys, and blood, result in relevant differences in organ absorbed doses when the radiotracers are applied for therapeutic purposes.
Collapse
Affiliation(s)
- T S T Damiana
- Department of Radiology & Nuclear Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - P Paraïso
- Department of Radiology & Nuclear Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - C de Ridder
- Department of Experimental Urology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - D Stuurman
- Department of Radiology & Nuclear Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Y Seimbille
- Department of Radiology & Nuclear Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - S U Dalm
- Department of Radiology & Nuclear Medicine, Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
36
|
Aggarwal M, Striegel DA, Hara M, Periwal V. Geometric and topological characterization of the cytoarchitecture of islets of Langerhans. PLoS Comput Biol 2023; 19:e1011617. [PMID: 37943957 PMCID: PMC10662755 DOI: 10.1371/journal.pcbi.1011617] [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: 05/11/2023] [Revised: 11/21/2023] [Accepted: 10/19/2023] [Indexed: 11/12/2023] Open
Abstract
The islets of Langerhans are critical endocrine micro-organs that secrete hormones regulating energy metabolism in animals. Insulin and glucagon, secreted by beta and alpha cells, respectively, are responsible for metabolic switching between fat and glucose utilization. Dysfunction in their secretion and/or counter-regulatory influence leads to diabetes. Debate in the field centers on the cytoarchitecture of islets, as the signaling that governs hormonal secretion depends on structural and functional factors, including electrical connectivity, innervation, vascularization, and physical proximity. Much effort has therefore been devoted to elucidating which architectural features are significant for function and how derangements in these features are correlated or causative for dysfunction, especially using quantitative network science or graph theory characterizations. Here, we ask if there are non-local features in islet cytoarchitecture, going beyond standard network statistics, that are relevant to islet function. An example is ring structures, or cycles, of α and δ cells surrounding β cell clusters or the opposite, β cells surrounding α and δ cells. These could appear in two-dimensional islet section images if a sphere consisting of one cell type surrounds a cluster of another cell type. To address these issues, we developed two independent computational approaches, geometric and topological, for such characterizations. For the latter, we introduce an application of topological data analysis to determine locations of topological features that are biologically significant. We show that both approaches, applied to a large collection of islet sections, are in complete agreement in the context both of developmental and diabetes-related changes in islet characteristics. The topological approach can be applied to three-dimensional imaging data for islets as well.
Collapse
Affiliation(s)
- Manu Aggarwal
- Laboratory of Biological Modeling, NIDDK, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Deborah A. Striegel
- Laboratory of Biological Modeling, NIDDK, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Manami Hara
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, The University of Chicago, Chicago, Illinois, United States of America
| | - Vipul Periwal
- Laboratory of Biological Modeling, NIDDK, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
37
|
Skelin Klemen M, Kopecky J, Dolenšek J, Stožer A. Human Beta Cell Functional Adaptation and Dysfunction in Insulin Resistance and Its Reversibility. Nephron Clin Pract 2023; 148:78-84. [PMID: 37883937 PMCID: PMC10860743 DOI: 10.1159/000534667] [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: 03/31/2023] [Accepted: 10/07/2023] [Indexed: 10/28/2023] Open
Abstract
BACKGROUND Beta cells play a key role in the pathophysiology of diabetes since their functional adaptation is able to maintain euglycemia in the face of insulin resistance, and beta cell decompensation or dysfunction is a necessary condition for full-blown type 2 diabetes (T2D). The mechanisms behind compensation and decompensation are incompletely understood, especially for human beta cells, and even less is known about influences of chronic kidney disease (CKD) or immunosupressive therapy after transplantation on these processes and the development of posttransplant diabetes. SUMMARY During compensation, beta cell sensitivity to glucose becomes left-shifted, i.e., their sensitivity to stimulation increases, and this is accompanied by enhanced signals along the stimulus-secretion coupling cascade from membrane depolarization to intracellular calcium and the most distal insulin secretion dynamics. There is currently no clear evidence regarding changes in intercellular coupling during this stage of disease progression. During decompensation, intracellular stimulus-secretion coupling remains enhanced to some extent at low or basal glucose concentrations but seems to become unable to generate effective signals to stimulate insulin secretion at high or otherwise stimulatory glucose concentrations. Additionally, intercellular coupling becomes disrupted, lowering the number of cells that contribute to secretion. During progression of CKD, beta cells also seem to drift from a compensatory left-shift to failure, and immunosupressants can further impair beta cell function following kidney transplantation. KEY MESSAGES Beta cell stimulus-secretion coupling is enhanced in compensated insulin resistance. With worsening insulin resistance, both intra- and intercellular coupling become disrupted. CKD can progressively disrupt beta cell function, but further studies are needed, especially regarding changes in intercellular coupling.
Collapse
Affiliation(s)
- Maša Skelin Klemen
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia,
| | - Jan Kopecky
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Jurij Dolenšek
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
| | - Andraž Stožer
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| |
Collapse
|
38
|
Minati MA, Fages A, Dauguet N, Zhu J, Jacquemin P. Optimized nucleus isolation protocol from frozen mouse tissues for single nucleus RNA sequencing application. Front Cell Dev Biol 2023; 11:1243863. [PMID: 37842081 PMCID: PMC10575574 DOI: 10.3389/fcell.2023.1243863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/12/2023] [Indexed: 10/17/2023] Open
Abstract
The single cell RNA sequencing technique has been particularly used during the last years, allowing major discoveries. However, the widespread application of this analysis has showed limitations. Indeed, the direct study of fresh tissues is not always feasible, notably in the case of genetically engineered mouse embryo or sensitive tissues whose integrity is affected by classical digestion methods. To overcome these limitations, single nucleus RNA sequencing offers the possibility to work with frozen samples. Thus, single nucleus RNA sequencing can be performed after genotyping-based selection on samples stocked in tissue bank and is applicable to retrospective studies. Therefore, this technique opens the field to a wide range of applications requiring adapted protocols for nucleus isolation according to the tissue considered. Here we developed a protocol of nucleus isolation from frozen murine placenta and pancreas. These two complex tissues were submitted to a combination of enzymatic and manual dissociation before undergoing different steps of washing and centrifugation. The entire protocol was performed with products usually present in a research lab. Before starting the sequencing process, nuclei were sorted by flow cytometry. The results obtained validate the efficiency of this protocol which is easy to set up and does not require the use of commercial kits. This specificity makes it adaptable to different organs and species. The association of this protocol with single nucleus RNA sequencing allows the study of complex samples that resist classical lysis methods due to the presence of fibrotic or fatty tissue, such as fibrotic kidney, tumors, embryonic tissues or fatty pancreas.
Collapse
Affiliation(s)
| | - Angeline Fages
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Nicolas Dauguet
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
- Flow Cytometry and Cell Sorting Facility (CYTF), de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Jingjing Zhu
- Ludwig Institute for Cancer Research, Université Catholique de Louvain, Brussels, Belgium
| | - Patrick Jacquemin
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
39
|
Pettway YD, Saunders DC, Brissova M. The human α cell in health and disease. J Endocrinol 2023; 258:e220298. [PMID: 37114672 PMCID: PMC10428003 DOI: 10.1530/joe-22-0298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 04/27/2023] [Indexed: 04/29/2023]
Abstract
In commemoration of 100 years since the discovery of glucagon, we review current knowledge about the human α cell. Alpha cells make up 30-40% of human islet endocrine cells and play a major role in regulating whole-body glucose homeostasis, largely through the direct actions of their main secretory product - glucagon - on peripheral organs. Additionally, glucagon and other secretory products of α cells, namely acetylcholine, glutamate, and glucagon-like peptide-1, have been shown to play an indirect role in the modulation of glucose homeostasis through autocrine and paracrine interactions within the islet. Studies of glucagon's role as a counterregulatory hormone have revealed additional important functions of the α cell, including the regulation of multiple aspects of energy metabolism outside that of glucose. At the molecular level, human α cells are defined by the expression of conserved islet-enriched transcription factors and various enriched signature genes, many of which have currently unknown cellular functions. Despite these common threads, notable heterogeneity exists amongst human α cell gene expression and function. Even greater differences are noted at the inter-species level, underscoring the importance of further study of α cell physiology in the human context. Finally, studies on α cell morphology and function in type 1 and type 2 diabetes, as well as other forms of metabolic stress, reveal a key contribution of α cell dysfunction to dysregulated glucose homeostasis in disease pathogenesis, making targeting the α cell an important focus for improving treatment.
Collapse
Affiliation(s)
- Yasminye D. Pettway
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, 37232, USA
| | - Diane C. Saunders
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, USA
| | - Marcela Brissova
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, USA
| |
Collapse
|
40
|
Sugawara H, Imai J, Yamamoto J, Izumi T, Kawana Y, Endo A, Kohata M, Seike J, Kubo H, Komamura H, Munakata Y, Asai Y, Hosaka S, Sawada S, Kodama S, Takahashi K, Kaneko K, Katagiri H. A highly sensitive strategy for monitoring real-time proliferation of targeted cell types in vivo. Nat Commun 2023; 14:3253. [PMID: 37316473 DOI: 10.1038/s41467-023-38897-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 05/22/2023] [Indexed: 06/16/2023] Open
Abstract
Cell proliferation processes play pivotal roles in timely adaptation to many biological situations. Herein, we establish a highly sensitive and simple strategy by which time-series showing the proliferation of a targeted cell type can be quantitatively monitored in vivo in the same individuals. We generate mice expressing a secreted type of luciferase only in cells producing Cre under the control of the Ki67 promoter. Crossing these with tissue-specific Cre-expressing mice allows us to monitor the proliferation time course of pancreatic β-cells, which are few in number and weakly proliferative, by measuring plasma luciferase activity. Physiological time courses, during obesity development, pregnancy and juvenile growth, as well as diurnal variation, of β-cell proliferation, are clearly detected. Moreover, this strategy can be utilized for highly sensitive ex vivo screening for proliferative factors for targeted cells. Thus, these technologies may contribute to advancements in broad areas of biological and medical research.
Collapse
Affiliation(s)
- Hiroto Sugawara
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Junta Imai
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Junpei Yamamoto
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomohito Izumi
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yohei Kawana
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akira Endo
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masato Kohata
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Junro Seike
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Haremaru Kubo
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroshi Komamura
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yuichiro Munakata
- Division of Metabolism and Diabetes, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Yoichiro Asai
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shinichiro Hosaka
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shojiro Sawada
- Division of Metabolism and Diabetes, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Shinjiro Kodama
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kei Takahashi
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Keizo Kaneko
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hideki Katagiri
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
41
|
Aldous N, Moin ASM, Abdelalim EM. Pancreatic β-cell heterogeneity in adult human islets and stem cell-derived islets. Cell Mol Life Sci 2023; 80:176. [PMID: 37270452 DOI: 10.1007/s00018-023-04815-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/27/2023] [Accepted: 05/19/2023] [Indexed: 06/05/2023]
Abstract
Recent studies reported that pancreatic β-cells are heterogeneous in terms of their transcriptional profiles and their abilities for insulin secretion. Sub-populations of pancreatic β-cells have been identified based on the functionality and expression of specific surface markers. Under diabetes condition, β-cell identity is altered leading to different β-cell sub-populations. Furthermore, cell-cell contact between β-cells and other endocrine cells within the islet play an important role in regulating insulin secretion. This highlights the significance of generating a cell product derived from stem cells containing β-cells along with other major islet cells for treating patients with diabetes, instead of transplanting a purified population of β-cells. Another key question is how close in terms of heterogeneity are the islet cells derived from stem cells? In this review, we summarize the heterogeneity in islet cells of the adult pancreas and those generated from stem cells. In addition, we highlight the significance of this heterogeneity in health and disease conditions and how this can be used to design a stem cell-derived product for diabetes cell therapy.
Collapse
Affiliation(s)
- Noura Aldous
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation, Education City, Doha, Qatar
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Education City, PO Box 34110, Doha, Qatar
| | - Abu Saleh Md Moin
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Education City, PO Box 34110, Doha, Qatar
- Research Department, Royal College of Surgeons in Ireland Bahrain, Adliya, Kingdom of Bahrain
| | - Essam M Abdelalim
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation, Education City, Doha, Qatar.
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Education City, PO Box 34110, Doha, Qatar.
| |
Collapse
|
42
|
Giarrizzo M, LaComb JF, Bialkowska AB. The Role of Krüppel-like Factors in Pancreatic Physiology and Pathophysiology. Int J Mol Sci 2023; 24:ijms24108589. [PMID: 37239940 DOI: 10.3390/ijms24108589] [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: 04/13/2023] [Revised: 05/04/2023] [Accepted: 05/06/2023] [Indexed: 05/28/2023] Open
Abstract
Krüppel-like factors (KLFs) belong to the family of transcription factors with three highly conserved zinc finger domains in the C-terminus. They regulate homeostasis, development, and disease progression in many tissues. It has been shown that KLFs play an essential role in the endocrine and exocrine compartments of the pancreas. They are necessary to maintain glucose homeostasis and have been implicated in the development of diabetes. Furthermore, they can be a vital tool in enabling pancreas regeneration and disease modeling. Finally, the KLF family contains proteins that act as tumor suppressors and oncogenes. A subset of members has a biphasic function, being upregulated in the early stages of oncogenesis and stimulating its progression and downregulated in the late stages to allow for tumor dissemination. Here, we describe KLFs' function in pancreatic physiology and pathophysiology.
Collapse
Affiliation(s)
- Michael Giarrizzo
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794, USA
| | - Joseph F LaComb
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794, USA
| | - Agnieszka B Bialkowska
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
43
|
Woeste MR, Shrestha R, Geller AE, Li S, Montoya-Durango D, Ding C, Hu X, Li H, Puckett A, Mitchell RA, Hayat T, Tan M, Li Y, McMasters KM, Martin RCG, Yan J. Irreversible electroporation augments β-glucan induced trained innate immunity for the treatment of pancreatic ductal adenocarcinoma. J Immunother Cancer 2023; 11:e006221. [PMID: 37072351 PMCID: PMC10124260 DOI: 10.1136/jitc-2022-006221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2023] [Indexed: 04/20/2023] Open
Abstract
BACKGROUND Pancreatic cancer (PC) is a challenging diagnosis that is yet to benefit from the advancements in immuno-oncologic treatments. Irreversible electroporation (IRE), a non-thermal method of tumor ablation, is used in treatment of select patients with locally-advanced unresectable PC and has potentiated the effect of certain immunotherapies. Yeast-derived particulate β-glucan induces trained innate immunity and successfully reduces murine PC tumor burden. This study tests the hypothesis that IRE may augment β-glucan induced trained immunity in the treatment of PC. METHODS β-Glucan-trained pancreatic myeloid cells were evaluated ex vivo for trained responses and antitumor function after exposure to ablated and unablated tumor-conditioned media. β-Glucan and IRE combination therapy was tested in an orthotopic murine PC model in wild-type and Rag-/- mice. Tumor immune phenotypes were assessed by flow cytometry. Effect of oral β-glucan in the murine pancreas was evaluated and used in combination with IRE to treat PC. The peripheral blood of patients with PC taking oral β-glucan after IRE was evaluated by mass cytometry. RESULTS IRE-ablated tumor cells elicited a potent trained response ex vivo and augmented antitumor functionality. In vivo, β-glucan in combination with IRE reduced local and distant tumor burden prolonging survival in a murine orthotopic PC model. This combination augmented immune cell infiltration to the PC tumor microenvironment and potentiated the trained response from tumor-infiltrating myeloid cells. The antitumor effect of this dual therapy occurred independent of the adaptive immune response. Further, orally administered β-glucan was identified as an alternative route to induce trained immunity in the murine pancreas and prolonged PC survival in combination with IRE. β-Glucan in vitro treatment also induced trained immunity in peripheral blood monocytes obtained from patients with treatment-naïve PC. Finally, orally administered β-glucan was found to significantly alter the innate cell landscape within the peripheral blood of five patients with stage III locally-advanced PC who had undergone IRE. CONCLUSIONS These data highlight a relevant and novel application of trained immunity within the setting of surgical ablation that may stand to benefit patients with PC.
Collapse
Affiliation(s)
- Matthew R Woeste
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, USA
- Division of Immunotherapy, The Hiram C. Polk Jr., MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Rejeena Shrestha
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, USA
- Division of Immunotherapy, The Hiram C. Polk Jr., MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Anne E Geller
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, USA
- Division of Immunotherapy, The Hiram C. Polk Jr., MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Shu Li
- Division of Immunotherapy, The Hiram C. Polk Jr., MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Diego Montoya-Durango
- Division of Immunotherapy, The Hiram C. Polk Jr., MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Chuanlin Ding
- Division of Immunotherapy, The Hiram C. Polk Jr., MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Xiaoling Hu
- Division of Immunotherapy, The Hiram C. Polk Jr., MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Hong Li
- Functional Immunomics Core, Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Aaron Puckett
- Functional Immunomics Core, Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Robert A Mitchell
- Division of Immunotherapy, The Hiram C. Polk Jr., MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Traci Hayat
- Division of Surgical Oncology, The Hiram C. Polk Jr., MD Department of Surgery, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Min Tan
- Division of Surgical Oncology, The Hiram C. Polk Jr., MD Department of Surgery, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Yan Li
- Division of Surgical Oncology, The Hiram C. Polk Jr., MD Department of Surgery, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Kelly M McMasters
- Division of Surgical Oncology, The Hiram C. Polk Jr., MD Department of Surgery, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Robert C G Martin
- Division of Surgical Oncology, The Hiram C. Polk Jr., MD Department of Surgery, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Jun Yan
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, USA
- Division of Immunotherapy, The Hiram C. Polk Jr., MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
| |
Collapse
|
44
|
Halperin R, Arnon L, Eden-Friedman Y, Tirosh A. Unique Characteristics of Patients with Von Hippel-Lindau Disease Defined by Various Diagnostic Criteria. Cancers (Basel) 2023; 15:cancers15061657. [PMID: 36980542 PMCID: PMC10046302 DOI: 10.3390/cancers15061657] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/28/2023] [Accepted: 03/05/2023] [Indexed: 03/30/2023] Open
Abstract
Von Hippel-Lindau (VHL) disease diagnosis is based on two criteria sets: International criteria (IC, two hemangioblastomas, one hemangioblastoma plus one visceral lesion, or VHL family history/pathogenic variant plus hemangioblastoma/visceral lesion); or Danish criteria (DC, two clinical manifestations, or VHL family history/pathogenic variant plus hemangioblastoma/visceral lesion). We aimed to compare the characteristics of patients with VHL-related pancreatic neuroendocrine tumor (vPNET) meeting either the clinical Danish criteria only (DOC) or IC to those with sporadic PNET (sPNET). The cohort included 33 patients with VHL (20 vPNETs) and 65 with sPNET. In terms of genetic testing and family history of VHL, 90.0% of the patients with vPNET in the IC group had a germline VHL pathogenic variant, and 70.0% had a family history of VHL vs. 20% and 10% in the DOC group, respectively (p < 0.05 for both). Patients with vPNET were younger at diagnosis compared with sPNET (51.6 ± 4.1 vs. 62.8 ± 1.5 years, p < 0.05). Patients in the IC group were younger at diagnosis with VHL, vPNET, pheochromocytoma, or paraganglioma (PPGL) and renal-cell carcinoma (RCC) than those in the DOC group (p < 0.05 for all comparisons). The most prevalent presenting manifestations were hemangioblastoma (42.8%) and PPGL (33.3%) vs. RCC (58.3%) and PNET (41.7%) in the IC vs. DOC groups. In conclusion, patients with vPNET meeting DOC criteria show greater similarity to sPNET. We suggest performing genetic testing, rather than solely using clinical criteria, for establishing the diagnosis of VHL.
Collapse
Affiliation(s)
- Reut Halperin
- ENTIRE Endocrine Neoplasia Translational Research Center, Sheba Rd. 2, Ramat Gan 6562601, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Division of Endocrinology, Diabetes and Metabolism, Sheba Medical Center, Ramat Gan 5266202, Israel
| | - Liat Arnon
- ENTIRE Endocrine Neoplasia Translational Research Center, Sheba Rd. 2, Ramat Gan 6562601, Israel
- Division of Endocrinology, Diabetes and Metabolism, Sheba Medical Center, Ramat Gan 5266202, Israel
| | - Yehudit Eden-Friedman
- Division of Endocrinology, Diabetes and Metabolism, Sheba Medical Center, Ramat Gan 5266202, Israel
| | - Amit Tirosh
- ENTIRE Endocrine Neoplasia Translational Research Center, Sheba Rd. 2, Ramat Gan 6562601, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Division of Endocrinology, Diabetes and Metabolism, Sheba Medical Center, Ramat Gan 5266202, Israel
| |
Collapse
|
45
|
Šterk M, Dolenšek J, Skelin Klemen M, Križančić Bombek L, Paradiž Leitgeb E, Kerčmar J, Perc M, Slak Rupnik M, Stožer A, Gosak M. Functional characteristics of hub and wave-initiator cells in β cell networks. Biophys J 2023; 122:784-801. [PMID: 36738106 PMCID: PMC10027448 DOI: 10.1016/j.bpj.2023.01.039] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 12/22/2022] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Islets of Langerhans operate as multicellular networks in which several hundred β cells work in synchrony to produce secretory pulses of insulin, a hormone crucial for controlling metabolic homeostasis. Their collective rhythmic activity is facilitated by gap junctional coupling and affected by their functional heterogeneity, but the details of this robust and coordinated behavior are still not fully understood. Recent advances in multicellular imaging and optogenetic and photopharmacological strategies, as well as in network science, have led to the discovery of specialized β cell subpopulations that were suggested to critically determine the collective dynamics in the islets. In particular hubs, i.e., β cells with many functional connections, are believed to significantly enhance communication capacities of the intercellular network and facilitate an efficient spreading of intercellular Ca2+ waves, whereas wave-initiator cells trigger intercellular signals in their cohorts. Here, we determined Ca2+ signaling characteristics of these two β cell subpopulations and the relationship between them by means of functional multicellular Ca2+ imaging in mouse pancreatic tissue slices in combination with methods of complex network theory. We constructed network layers based on individual Ca2+ waves to identify wave initiators, and functional correlation-based networks to detect hubs. We found that both cell types exhibit a higher-than-average active time under both physiological and supraphysiological glucose concentrations, but also that they differ significantly in many other functional characteristics. Specifically, Ca2+ oscillations in hubs are more regular, and their role appears to be much more stable over time than for initiator cells. Moreover, in contrast to wave initiators, hubs transmit intercellular signals faster than other cells, which implies a stronger intercellular coupling. Our research indicates that hubs and wave-initiator cell subpopulations are both natural features of healthy pancreatic islets, but their functional roles in principle do not overlap and should thus not be considered equal.
Collapse
Affiliation(s)
- Marko Šterk
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia; Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Jurij Dolenšek
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia; Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | | | | | | | - Jasmina Kerčmar
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Matjaž Perc
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan; Alma Mater Europaea, Maribor, Slovenia; Complexity Science Hub Vienna, Vienna, Austria; Department of Physics, Kyung Hee University, Dongdaemun-gu, Seoul, Republic of Korea
| | - Marjan Slak Rupnik
- Faculty of Medicine, University of Maribor, Maribor, Slovenia; Alma Mater Europaea, Maribor, Slovenia; Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Andraž Stožer
- Faculty of Medicine, University of Maribor, Maribor, Slovenia.
| | - Marko Gosak
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia; Faculty of Medicine, University of Maribor, Maribor, Slovenia; Alma Mater Europaea, Maribor, Slovenia.
| |
Collapse
|
46
|
Richardson TM, Saunders DC, Haliyur R, Shrestha S, Cartailler JP, Reinert RB, Petronglo J, Bottino R, Aramandla R, Bradley AM, Jenkins R, Phillips S, Kang H, Caicedo A, Powers AC, Brissova M. Human pancreatic capillaries and nerve fibers persist in type 1 diabetes despite beta cell loss. Am J Physiol Endocrinol Metab 2023; 324:E251-E267. [PMID: 36696598 PMCID: PMC10027091 DOI: 10.1152/ajpendo.00246.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/26/2023]
Abstract
The autonomic nervous system regulates pancreatic function. Islet capillaries are essential for the extension of axonal projections into islets, and both of these structures are important for appropriate islet hormone secretion. Because beta cells provide important paracrine cues for islet glucagon secretion and neurovascular development, we postulated that beta cell loss in type 1 diabetes (T1D) would lead to a decline in intraislet capillaries and reduction of islet innervation, possibly contributing to abnormal glucagon secretion. To define morphological characteristics of capillaries and nerve fibers in islets and acinar tissue compartments, we analyzed neurovascular assembly across the largest cohort of T1D and normal individuals studied thus far. Because innervation has been studied extensively in rodent models of T1D, we also compared the neurovascular architecture between mouse and human pancreas and assembled transcriptomic profiles of molecules guiding islet angiogenesis and neuronal development. We found striking interspecies differences in islet neurovascular assembly but relatively modest differences at transcriptome level, suggesting that posttranscriptional regulation may be involved in this process. To determine whether islet neurovascular arrangement is altered after beta cell loss in T1D, we compared pancreatic tissues from non-diabetic, recent-onset T1D (<10-yr duration), and longstanding T1D (>10-yr duration) donors. Recent-onset T1D showed greater islet and acinar capillary density compared to non-diabetic and longstanding T1D donors. Both recent-onset and longstanding T1D had greater islet nerve fiber density compared to non-diabetic donors. We did not detect changes in sympathetic axons in either T1D cohort. Additionally, nerve fibers overlapped with extracellular matrix (ECM), supporting its role in the formation and function of axonal processes. These results indicate that pancreatic capillaries and nerve fibers persist in T1D despite beta cell loss, suggesting that alpha cell secretory changes may be decoupled from neurovascular components.NEW & NOTEWORTHY Defining the neurovascular architecture in the pancreas of individuals with type 1 diabetes (T1D) is crucial to understanding the mechanisms of dysregulated glucagon secretion. In the largest T1D cohort of biobanked tissues analyzed to date, we found that pancreatic capillaries and nerve fibers persist in human T1D despite beta cell loss, suggesting that alpha cell secretory changes may be decoupled from neurovascular components. Because innervation has been studied extensively in rodent T1D models, our studies also provide the first rigorous direct comparisons of neurovascular assembly in mouse and human, indicating dramatic interspecies differences.
Collapse
Affiliation(s)
- Tiffany M Richardson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Diane C Saunders
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Rachana Haliyur
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan, United States
| | - Shristi Shrestha
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Creative Data Solutions, Vanderbilt Center for Stem Cell Biology, Nashville, Tennessee, United States
| | - Jean-Philippe Cartailler
- Creative Data Solutions, Vanderbilt Center for Stem Cell Biology, Nashville, Tennessee, United States
| | - Rachel B Reinert
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, United States
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Jenna Petronglo
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Rita Bottino
- Imagine Pharma, Pittsburgh, Pennsylvania, United States
| | - Radhika Aramandla
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Amber M Bradley
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Regina Jenkins
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Sharon Phillips
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Hakmook Kang
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Alejandro Caicedo
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, United States
- Program of Neuroscience, University of Miami Miller School of Medicine, Miami, Florida, United States
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Alvin C Powers
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Department of Veterans Affairs Tennessee Valley Healthcare, Nashville, Tennessee, United States
| | - Marcela Brissova
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| |
Collapse
|
47
|
De la Cruz-Concepción B, Flores-Cortez YA, Barragán-Bonilla MI, Mendoza-Bello JM, Espinoza-Rojo M. Insulin: A connection between pancreatic β cells and the hypothalamus. World J Diabetes 2023; 14:76-91. [PMID: 36926659 PMCID: PMC10011898 DOI: 10.4239/wjd.v14.i2.76] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/13/2022] [Accepted: 01/17/2023] [Indexed: 02/14/2023] Open
Abstract
Insulin is a hormone secreted by pancreatic β cells. The concentration of glucose in circulation is proportional to the secretion of insulin by these cells. In target cells, insulin binds to its receptors and activates phosphatidylinositol-3-kinase/protein kinase B, inducing different mechanisms depending on the cell type. In the liver it activates the synthesis of glycogen, in adipose tissue and muscle it allows the capture of glucose, and in the hypothalamus, it regulates thermogenesis and appetite. Defects in insulin function [insulin resistance (IR)] are related to the development of neurodegenerative diseases in obese people. Furthermore, in obesity and diabetes, its role as an anorexigenic hormone in the hypothalamus is diminished during IR. Therefore, hyperphagia prevails, which aggravates hyper-glycemia and IR further, becoming a vicious circle in which the patient cannot regulate their need to eat. Uncontrolled calorie intake induces an increase in reactive oxygen species, overcoming cellular antioxidant defenses (oxidative stress). Reactive oxygen species activate stress-sensitive kinases, such as c-Jun N-terminal kinase and p38 mitogen-activated protein kinase, that induce phos-phorylation in serine residues in the insulin receptor, which blocks the insulin signaling pathway, continuing the mechanism of IR. The brain and pancreas are organs mainly affected by oxidative stress. The use of drugs that regulate food intake and improve glucose metabolism is the conventional therapy to improve the quality of life of these patients. Currently, the use of antioxidants that regulate oxidative stress has given good results because they reduce oxidative stress and inflammatory processes, and they also have fewer side effects than synthetic drugs.
Collapse
Affiliation(s)
- Brenda De la Cruz-Concepción
- Molecular and Genomic Biology Laboratory, Faculty of Chemical-Biological Sciences, Autonomous University of Guerrero, Chilpancingo 39070, Guerrero, Mexico
| | - Yaccil Adilene Flores-Cortez
- Molecular and Genomic Biology Laboratory, Faculty of Chemical-Biological Sciences, Autonomous University of Guerrero, Chilpancingo 39070, Guerrero, Mexico
| | - Martha Isela Barragán-Bonilla
- Molecular and Genomic Biology Laboratory, Faculty of Chemical-Biological Sciences, Autonomous University of Guerrero, Chilpancingo 39070, Guerrero, Mexico
| | - Juan Miguel Mendoza-Bello
- Molecular and Genomic Biology Laboratory, Faculty of Chemical-Biological Sciences, Autonomous University of Guerrero, Chilpancingo 39070, Guerrero, Mexico
| | - Monica Espinoza-Rojo
- Molecular and Genomic Biology Laboratory, Faculty of Chemical-Biological Sciences, Autonomous University of Guerrero, Chilpancingo 39070, Guerrero, Mexico
| |
Collapse
|
48
|
Homma J, Sekine H, Shimizu T. Tricultured Cell Sheets Develop into Functional Pancreatic Islet Tissue with a Vascular Network. Tissue Eng Part A 2023; 29:211-224. [PMID: 36565034 DOI: 10.1089/ten.tea.2022.0167] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Methods to induce islet β-cells from induced pluripotent stem cells or embryonic stem cells have been established. However, islet β-cells are susceptible to apoptosis under hypoxic conditions, so the technique used to transplant β-cells must maintain the viability of cells in vivo. This study describes the development of a tricultured cell sheet, which was made by coculturing islet β-cells, vascular endothelial cells, and mesenchymal stem cells for 1 day. The islet β-cells in the tricultured cell sheet self-organized into islet-like structures surrounded by a dense vascular network in vitro. Triple-layered tricultured cell sheets engrafted well after transplantation in vivo and developed into insulin-secreting tissue with abundant blood vessels and a high density of islet β-cells. We anticipate that the tricultured cell sheet could be used as an in vitro pseudo-islet model for pharmaceutical testing and may have potential for development into transplantable grafts for use in regenerative medicine.
Collapse
Affiliation(s)
- Jun Homma
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, Japan
| | - Hidekazu Sekine
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, Japan
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
49
|
Melamed JR, Yerneni SS, Arral ML, LoPresti ST, Chaudhary N, Sehrawat A, Muramatsu H, Alameh MG, Pardi N, Weissman D, Gittes GK, Whitehead KA. Ionizable lipid nanoparticles deliver mRNA to pancreatic β cells via macrophage-mediated gene transfer. SCIENCE ADVANCES 2023; 9:eade1444. [PMID: 36706177 PMCID: PMC9882987 DOI: 10.1126/sciadv.ade1444] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 12/27/2022] [Indexed: 05/19/2023]
Abstract
Systemic messenger RNA (mRNA) delivery to organs outside the liver, spleen, and lungs remains challenging. To overcome this issue, we hypothesized that altering nanoparticle chemistry and administration routes may enable mRNA-induced protein expression outside of the reticuloendothelial system. Here, we describe a strategy for delivering mRNA potently and specifically to the pancreas using lipid nanoparticles. Our results show that delivering lipid nanoparticles containing cationic helper lipids by intraperitoneal administration produces robust and specific protein expression in the pancreas. Most resultant protein expression occurred within insulin-producing β cells. Last, we found that pancreatic mRNA delivery was dependent on horizontal gene transfer by peritoneal macrophage exosome secretion, an underappreciated mechanism that influences the delivery of mRNA lipid nanoparticles. We anticipate that this strategy will enable gene therapies for intractable pancreatic diseases such as diabetes and cancer.
Collapse
Affiliation(s)
- Jilian R. Melamed
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Mariah L. Arral
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Samuel T. LoPresti
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Namit Chaudhary
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Anuradha Sehrawat
- Department of Pediatric Surgery, Department of Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Hiromi Muramatsu
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Norbert Pardi
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Drew Weissman
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - George K. Gittes
- Department of Pediatric Surgery, Department of Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Kathryn A. Whitehead
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| |
Collapse
|
50
|
Khomari F, Kiani B, Alizadeh-Fanalou S, Babaei M, Kalantari-Hesari A, Alipourfard I, Mirzaei F, Yarahmadi S, Bahreini E. Effectiveness of Hydroalcoholic Seed Extract of Securigera securidaca on Pancreatic Local Renin-Angiotensin System and Its Alternative Pathway in Streptozotocin-Induced Diabetic Animal Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:7285036. [PMID: 36647426 PMCID: PMC9840543 DOI: 10.1155/2023/7285036] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/17/2022] [Accepted: 12/22/2022] [Indexed: 01/09/2023]
Abstract
Background Available data suggest inhibition of the pancreatic local-renin-angiotensin system (RAS) reduces tissue complications of diabetes. The purpose of the present study was to investigate the effect of hydroalcoholic seed extract of Securigera securidaca (S. securidaca) (HESS) on the pancreatic local-RAS and its alternative pathway. Methods Three doses of HESS were orally administered to three groups of diabetic male Wistar rats, and the results were compared with both diabetic and healthy control groups. After 35 days of treatment, the groups were assessed for the levels of pancreatic local-RAS components, including renin, angiotensinogen, ACE, and Ang II, as well as ACE2 and Ang-(1-7) in the alternative pathway. The effect of herbal medicine treatment on tissue damage status was investigated by evaluating tissue levels of oxidative stress, proinflammatory and anti-inflammatory cytokines, and through histopathological examination of the pancreas. Results HESS showed a dose-dependent palliative effect on the tissue oxidative stress profile (P < 0.05) as well as the levels of pancreatic local-RAS components (P < 0.05), compared to diabetic control group. Considering the interrelationship between tissue oxidative stress and local-RAS activity, the moderating effect of HESS on this relationship could be attributed to the increase in total tissue antioxidant capacity (TAC) and pancreatic Ang-(1-7) concentration. Decrease in local-RAS activity was associated with decrease in the tissue levels of inflammatory cytokines (IL1, IL6, and TNFα) (P < 0.05) and increase in the levels of anti-inflammatory cytokine of IL-10 (P < 0.05). In addition, histological results were consistent with tissue biochemical results. Conclusions Due to the reduction of local pancreatic RAS activity as well as oxidative stress and proinflammatory cytokines following treatment with HESS, S. securidaca seed can be proposed as a suitable herbal supplement in the drug-treatment of diabetes.
Collapse
Affiliation(s)
- Fatemeh Khomari
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Bahar Kiani
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shahin Alizadeh-Fanalou
- Nephrology and Kidney Transplant center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Mohammad Babaei
- Department of Clinical Sciences, Faculty of Veterinary Science, Bu-Ali Sina University, Hamedan, Iran
| | - Ali Kalantari-Hesari
- Department of Clinical Sciences, Faculty of Veterinary Science, Bu-Ali Sina University, Hamedan, Iran
| | - Iraj Alipourfard
- Institute of Biology, Biotechnology and Environmental Protection, Faculty of Natural Sciences, University of Silesia, Bankowa 9, 40-007 Katow, Poland
| | - Fatemeh Mirzaei
- Department of Anatomical Sciences, School of Medicine, Hamedan University of Medical Sciences, Hamedan, Iran
| | - Sahar Yarahmadi
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Elham Bahreini
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|