1
|
He Z, Liu Q, Wang Y, Zhao B, Zhang L, Yang X, Wang Z. The role of endoplasmic reticulum stress in type 2 diabetes mellitus mechanisms and impact on islet function. PeerJ 2025; 13:e19192. [PMID: 40166045 PMCID: PMC11956770 DOI: 10.7717/peerj.19192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 02/26/2025] [Indexed: 04/02/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a globally prevalent metabolic disorder characterized by insulin resistance and dysfunction of islet cells. Endoplasmic reticulum (ER) stress plays a crucial role in the pathogenesis and progression of T2DM, especially in the function and survival of β-cells. β-cells are particularly sensitive to ER stress because they require substantial insulin synthesis and secretion energy. In the early stages of T2DM, the increased demand for insulin exacerbates β-cell ER stress. Although the unfolded protein response (UPR) can temporarily alleviate this stress, prolonged or excessive stress leads to pancreatic cell dysfunction and apoptosis, resulting in insufficient insulin secretion. This review explores the mechanisms of ER stress in T2DM, particularly its impact on islet cells. We discuss how ER stress activates UPR signaling pathways to regulate protein folding and degradation, but when stress becomes excessive, these pathways may contribute to β-cell death. A deeper understanding of how ER stress impacts islet cells could lead to the development of novel T2DM treatment strategies aimed at improving islet function and slowing disease progression.
Collapse
Affiliation(s)
- Zhaxicao He
- Gansu University of Chinese Medicine, Lanzhou, China
| | - Qian Liu
- Gansu University of Chinese Medicine, Lanzhou, China
| | - Yan Wang
- Gansu University of Chinese Medicine, Lanzhou, China
| | - Bing Zhao
- Gansu University of Chinese Medicine, Lanzhou, China
| | - Lumei Zhang
- Gansu University of Chinese Medicine, Lanzhou, China
| | - Xia Yang
- Tianshui Hospital of Traditional Chinese Medicine, Tianshui, China
| | - Zhigang Wang
- Gansu University of Chinese Medicine, Lanzhou, China
- Tianshui Hospital of Traditional Chinese Medicine, Tianshui, China
| |
Collapse
|
2
|
M Zand A, Anastassov S, Frei T, Khammash M. Multi-Layer Autocatalytic Feedback Enables Integral Control Amidst Resource Competition and Across Scales. ACS Synth Biol 2025. [PMID: 40116396 DOI: 10.1021/acssynbio.4c00575] [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: 03/23/2025]
Abstract
Integral feedback control strategies have proven effective in regulating protein expression in unpredictable cellular environments. These strategies, grounded in model-based designs and control theory, have advanced synthetic biology applications. Autocatalytic integral feedback controllers, utilizing positive autoregulation for integral action, are one class of simplest architectures to design integrators. This class of controllers offers unique features, such as robustness against dilution effects and cellular growth, as well as the potential for synthetic realizations across different biological scales, owing to their similarity to self-regenerative behaviors widely observed in nature. Despite this, their potential has not yet been fully exploited. One key reason, we discuss, is that their effectiveness is often hindered by resource competition and context-dependent couplings. This study addresses these challenges using a multilayer feedback strategy. Our designs enabled population-level integral feedback and multicellular integrators, where the control function emerges as a property of coordinated interactions distributed across different cell populations coexisting in a multicellular consortium. We provide a generalized mathematical framework for modeling resource competition in complex genetic networks, supporting the design of intracellular control circuits. The use of our proposed multilayer autocatalytic controllers is examined in two typical control tasks that pose significant relevance to synthetic biology applications: concentration regulation and ratiometric control. We define a ratiometric control task and solve it using a variant of our controller. The effectiveness of our controller motifs is demonstrated through a range of application examples, from precise regulation of gene expression and gene ratios in embedded designs to population growth and coculture composition control in multicellular designs within engineered microbial ecosystems. These findings offer a versatile approach to achieving robust adaptation and homeostasis from subcellular to multicellular scales.
Collapse
Affiliation(s)
- Armin M Zand
- ETH Zurich, Department of Biosystems Science and Engineering, Schanzenstrasse 44, Basel 4056, Switzerland
| | - Stanislav Anastassov
- ETH Zurich, Department of Biosystems Science and Engineering, Schanzenstrasse 44, Basel 4056, Switzerland
| | - Timothy Frei
- ETH Zurich, Department of Biosystems Science and Engineering, Schanzenstrasse 44, Basel 4056, Switzerland
| | - Mustafa Khammash
- ETH Zurich, Department of Biosystems Science and Engineering, Schanzenstrasse 44, Basel 4056, Switzerland
| |
Collapse
|
3
|
Nota G, Cimmino W, Singh S, Darwish IA, La Rocca C, Carbone F, Matarese G, Cinti S. A portable and ecological paper-based device for glucose monitoring in peripheral blood mononuclear cell lysates. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2025; 17:2529-2535. [PMID: 39976010 DOI: 10.1039/d5ay00057b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The increasing need for point-of-care (POC) testing has prompted a rise in the popularity of affordable biosensors that are eco-friendly, especially paper-based electrochemical sensors. This research introduces a biodegradable paper-based enzymatic biosensor for detecting glucose levels in intricate biological samples, such as cell lysates. This biosensor uses Prussian Blue (PB) as a mediator and glucose oxidase to detect glucose with excellent accuracy using direct electrochemical signals. Screen printing using Whatman filter paper produced a better biosensor than other substrates. The PB concentration of 12.5 mmol L-1 was found to be optimal and resulted in an operating potential of -0.1 V, which helped decrease interference from other active substances and improved its selectivity. Calibration was found to be linear up to a concentration of 2 mmol L-1 with a detection limit of 40 μmol L-1 and a limit of quantification of 120 μmol L-1. Moreover, experiments performed on cell lysates obtained from peripheral blood mononuclear cells (PBMCs) suggest the possible application of biosensors to measure glucose levels in vitro in both stimulated and unstimulated cells. This feature underscores its promise for use in monitoring metabolism and conducting diagnostic applications. The paper-based biosensor is an alternative to the current platform for the development of an eco-friendly, portable glucose-sensitive biosensor for point-of-care monitoring of glucose. Its flexibility and efficiency make it a strong candidate for use in the field of POC diagnostics, especially in areas of limited resources and in conditions where there is a problem with glucose dysregulation including diabetes and other related metabolic disorders.
Collapse
Affiliation(s)
- Grazia Nota
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131, Naples, Italy.
| | - Wanda Cimmino
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131, Naples, Italy.
| | - Sima Singh
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131, Naples, Italy.
| | - Ibrahim A Darwish
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P. O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Claudia La Rocca
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore", Consiglio Nazionale delle Ricerche (IEOS-CNR), Napoli, Italy
| | - Fortunata Carbone
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore", Consiglio Nazionale delle Ricerche (IEOS-CNR), Napoli, Italy
| | - Giuseppe Matarese
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore", Consiglio Nazionale delle Ricerche (IEOS-CNR), Napoli, Italy
- Treg Cell Lab, Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", Napoli, Italy
| | - Stefano Cinti
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131, Naples, Italy.
- Bioelectronics Task Force at University of Naples Federico II, Via Cinthia 21, 80126, Naples, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, USA
| |
Collapse
|
4
|
Thakur R, Rana S, Baltoo R. Exploring fluoride's role in diabetes development: A review. J Trace Elem Med Biol 2025; 89:127635. [PMID: 40132392 DOI: 10.1016/j.jtemb.2025.127635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/13/2025] [Accepted: 03/13/2025] [Indexed: 03/27/2025]
Abstract
PURPOSE The element fluorine, which is never found in nature in a free state, is the source of the fluoride ion. When fluoride intake is excessive, it can cause various impairments in living organism. This review aims to assess the relationship between fluoride exposure and glucose metabolism, considering positive, negative, and null findings, with a focus on its potential role in insulin resistance and diabetes-related complications. METHODS Numerous studies that have demonstrated changes in blood glucose and insulin variations due to fluoride are included in our analysis on the bases of their relevance. Twenty significant research papers from Pubmed, Google Scholar, and Research Gate are included up to January 2025 using search terms such as "Fluoride," "Toxicity," "Diabetes," "Insulin resistance," "fluoride and diabetes," "fluoride and insulin," "fluoride and blood glucose" in this review. Of the 20 research papers, 14 involve normal organisms unaffected by diabetes or complications connected to the disease, serving as standard animal models, while 5 involve animals exposed to diabetes and 1 is a human population study. RESULTS The findings suggest a negative association between fluoride exposure and diabetes, as studies indicate fluoride's potential role in impairing glucose homeostasis and increasing insulin resistance. These research studies showed how fluoride affected the participants' blood sugar and diabetes-related complications. CONCLUSION This study highlights how important it is to comprehend how fluoride may contribute to diabetes or diabetes-related complications, and it makes recommendations for future research directions that might lead to the discovery of efficient treatment measures to avoid them.
Collapse
Affiliation(s)
- Ruhi Thakur
- School of Biological and Environmental Sciences, Shoolini University, Solan, India.
| | - Srishti Rana
- School of Biological and Environmental Sciences, Shoolini University, Solan, India
| | - Rashi Baltoo
- School of Biological and Environmental Sciences, Shoolini University, Solan, India
| |
Collapse
|
5
|
Barakat H, Aljutaily T. Role of γ-Aminobutyric Acid (GABA) as an Inhibitory Neurotransmitter in Diabetes Management: Mechanisms and Therapeutic Implications. Biomolecules 2025; 15:399. [PMID: 40149935 PMCID: PMC11940341 DOI: 10.3390/biom15030399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/03/2025] [Accepted: 03/07/2025] [Indexed: 03/29/2025] Open
Abstract
GABA (γ-Aminobutyric Acid), a well-established inhibitory neurotransmitter in the central nervous system, has garnered considerable interest for its potential role in diabetes management, particularly due to its presence in pancreatic islets. This review aims to explore the therapeutic role of GABA in diabetes management and its potential mechanisms for antidiabetic effects. Relevant studies were searched across databases such as PubMed and ScienceDirect, applying strict eligibility criteria focused on GABA administration methods and diabetic models. The collective results showed that the administration of GABA in diabetic models resulted in remarkable enhancements in glucose and insulin homeostasis, favorable modifications in lipid profiles, and amelioration of dysfunctions across neural, hepatic, renal, and cardiac systems. The findings from the literature demonstrated that GABAergic signaling within pancreatic tissues can significantly contribute to the stimulation of β cell proliferation through the facilitation of a sustained trans-differentiation process, wherein glucagon-secreting α cells are converted into insulin-secreting β-like cells. In addition, activated GABAergic signaling can trigger the initiation of the PI3K/AKT signaling pathway within pancreatic tissues, leading to improved insulin signaling and maintained glucose homeostasis. GABAergic signaling can further function within hepatic tissues, promoting inhibitory effects on the expression of genes related to gluconeogenesis and lipogenesis. Moreover, GABA may enhance gut microbiota diversity by attenuating gut inflammation, attributable to its anti-inflammatory and immunomodulatory properties. Furthermore, the neuroprotective effects of GABA play a significant role in ameliorating neural disorders associated with diabetes by facilitating a substantial reduction in neuronal apoptosis. In conclusion, GABA emerges as a promising candidate for an antidiabetic agent; however, further research is highly encouraged to develop a rigorously designed framework that comprehensively identifies and optimizes the appropriate dosages and intervention methods for effectively managing and combating diabetes.
Collapse
Affiliation(s)
- Hassan Barakat
- Department of Food Science and Human Nutrition, College of Agriculture and Food, Qassim University, Buraydah 51452, Saudi Arabia;
| | | |
Collapse
|
6
|
Basher ARMA, Hallinan C, Lee K. Heterogeneity-Preserving Discriminative Feature Selection for Disease-Specific Subtype Discovery. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.05.14.540686. [PMID: 38187596 PMCID: PMC10769187 DOI: 10.1101/2023.05.14.540686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
The identification of disease-specific subtypes can provide valuable insights into disease progression and potential individualized therapies, important aspects of precision medicine given the complex nature of disease heterogeneity. The advent of high-throughput technologies has enabled the generation and analysis of various molecular data types, such as single-cell RNA-seq, proteomic, and imaging datasets, on a large scale. While these datasets offer opportunities for subtype discovery, they also pose challenges in finding subtype signatures due to their high dimensionality. Feature selection, a key step in the machine learning pipeline, involves selecting signatures that reduce feature size for more efficient downstream computational analysis. Although many existing methods focus on selecting features that differentiate known diseases or cell states, they often struggle to identify features that both preserve heterogeneity and reveal subtypes. To address this, we utilized deep metric learning-based feature embedding to explore the statistical properties of features crucial for preserving heterogeneity. Our analysis indicated that features with a notable difference in interquartile range (IQR) between classes hold important subtype information. Guided by this insight, we developed a statistical method called PHet (Preserving Heterogeneity), which employs iterative subsampling and differential analysis of IQR combined with Fisher's method to identify a small set of features that preserve heterogeneity and enhance subtype clustering quality. Validation on public single-cell RNA-seq and microarray datasets demonstrated PHet's ability to maintain sample heterogeneity while distinguishing known disease/cell states, with a tendency to outperform previous differential expression and outlier-based methods. Furthermore, an analysis of a single-cell RNA-seq dataset from mouse tracheal epithelial cells identified two distinct basal cell subtypes differentiating towards a luminal secretory phenotype using PHet-based features, demonstrating promising results in a real-data application. These results highlight PHet's potential to enhance our understanding of disease mechanisms and cell differentiation, contributing significantly to the field of personalized medicine.
Collapse
Affiliation(s)
- Abdur Rahman M A Basher
- Vascular Biology Program, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Caleb Hallinan
- Vascular Biology Program, Boston Children's Hospital, Boston, MA 02115, USA
| | - Kwonmoo Lee
- Vascular Biology Program, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
7
|
Lee WD, Weilandt DR, Liang L, MacArthur MR, Jaiswal N, Ong O, Mann CG, Chu Q, Hunter CJ, Ryseck RP, Lu W, Oschmann AM, Cowan AJ, TeSlaa TA, Bartman CR, Jang C, Baur JA, Titchenell PM, Rabinowitz JD. Lactate homeostasis is maintained through regulation of glycolysis and lipolysis. Cell Metab 2025; 37:758-771.e8. [PMID: 39889702 PMCID: PMC11926601 DOI: 10.1016/j.cmet.2024.12.009] [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: 04/30/2024] [Revised: 09/19/2024] [Accepted: 12/17/2024] [Indexed: 02/03/2025]
Abstract
Lactate is among the highest flux circulating metabolites. It is made by glycolysis and cleared by both tricarboxylic acid (TCA) cycle oxidation and gluconeogenesis. Severe lactate elevations are life-threatening, and modest elevations predict future diabetes. How lactate homeostasis is maintained, however, remains poorly understood. Here, we identify, in mice, homeostatic circuits regulating lactate production and consumption. Insulin induces lactate production by upregulating glycolysis. We find that hyperlactatemia inhibits insulin-induced glycolysis, thereby suppressing excess lactate production. Unexpectedly, insulin also promotes lactate TCA cycle oxidation. The mechanism involves lowering circulating fatty acids, which compete with lactate for mitochondrial oxidation. Similarly, lactate can promote its own consumption by lowering circulating fatty acids via the adipocyte-expressed G-protein-coupled receptor hydroxycarboxylic acid receptor 1 (HCAR1). Quantitative modeling suggests that these mechanisms suffice to produce lactate homeostasis, with robustness to noise and perturbation of individual regulatory mechanisms. Thus, through regulation of glycolysis and lipolysis, lactate homeostasis is maintained.
Collapse
Affiliation(s)
- Won Dong Lee
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Daniel R Weilandt
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Lingfan Liang
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Michael R MacArthur
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Natasha Jaiswal
- Department of Health and Kinesiology, Purdue University, West Lafayette, IN, USA
| | - Olivia Ong
- Department of Health and Kinesiology, Purdue University, West Lafayette, IN, USA
| | - Charlotte G Mann
- Department of Health Sciences and Technology, ETH Zürich, Schwerzenbach, Switzerland
| | - Qingwei Chu
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Craig J Hunter
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Rolf-Peter Ryseck
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Wenyun Lu
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Anna M Oschmann
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Alexis J Cowan
- Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Tara A TeSlaa
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Caroline R Bartman
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cholsoon Jang
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Joseph A Baur
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul M Titchenell
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joshua D Rabinowitz
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
8
|
Wong KK, Ab. Hamid SS. Multiomics in silico analysis identifies TM4SF4 as a cell surface target in hepatocellular carcinoma. PLoS One 2025; 20:e0307048. [PMID: 39999090 PMCID: PMC11856526 DOI: 10.1371/journal.pone.0307048] [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: 06/28/2024] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
The clinical application of cellular immunotherapy in hepatocellular carcinoma (HCC) is impeded by the lack of a cell surface target frequently expressed in HCC cells and with minimal presence in normal tissues to reduce on-target, off-tumor toxicity. To address this, an in silico multomics analysis was conducted to identify an optimal therapeutic target in HCC. A longlist of genes (n = 12,948) expressed in HCCs according to The Human Protein Atlas database were examined. Eight genes were shortlisted to identify one with the highest expression in HCCs, without being shed into circulation, and with restrictive expression profile in other normal human tissues. A total of eight genes were shortlisted and subsequently ranked according to the combination of their transcript and protein expression levels in HCC cases (n = 791) derived from four independent datasets. TM4SF4 was the top-ranked target with the highest expression in HCCs. TM4SF4 showed more favorable expression profile with significantly lower expression in normal human tissues but more highly expressed in HCC compared with seven other common HCC therapeutic targets. Furthermore, scRNA-seq and immunohistochemistry datasets showed that TM4SF4 was absent in immune cell populations but highly expressed in the bile duct canaliculi of hepatocytes, regions inaccessible to immune cells. In scRNA-seq dataset of HCCs, TM4SF4 expression was positively associated with mitochondrial components and oxidative phosphorylation Gene Ontologies in HCC cells (n = 15,787 cells), suggesting its potential roles in mitochondrial-mediated oncogenic effects in HCC. Taken together, TM4SF4 is proposed as a promising cell surface target in HCC due to its high expression in HCC cells with restricted expression profile in non-cancerous tissues, and association with HCC oncogenic pathways.
Collapse
Affiliation(s)
- Kah Keng Wong
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Suzina Sheikh Ab. Hamid
- Tissue Bank Unit, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
- Department of Otorhinolaryngology-Head & Neck Surgery, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
9
|
Hu C, Chen Y, Yin X, Xu R, Yin C, Wang C, Zhao Y. Pancreatic endocrine and exocrine signaling and crosstalk in physiological and pathological status. Signal Transduct Target Ther 2025; 10:39. [PMID: 39948335 PMCID: PMC11825823 DOI: 10.1038/s41392-024-02098-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/20/2024] [Accepted: 12/03/2024] [Indexed: 02/16/2025] Open
Abstract
The pancreas, an organ with dual functions, regulates blood glucose levels through the endocrine system by secreting hormones such as insulin and glucagon. It also aids digestion through the exocrine system by secreting digestive enzymes. Complex interactions and signaling mechanisms between the endocrine and exocrine functions of the pancreas play a crucial role in maintaining metabolic homeostasis and overall health. Compelling evidence indicates direct and indirect crosstalk between the endocrine and exocrine parts, influencing the development of diseases affecting both. From a developmental perspective, the exocrine and endocrine parts share the same origin-the "tip-trunk" domain. In certain circumstances, pancreatic exocrine cells may transdifferentiate into endocrine-like cells, such as insulin-secreting cells. Additionally, several pancreatic diseases, including pancreatic cancer, pancreatitis, and diabetes, exhibit potential relevance to both endocrine and exocrine functions. Endocrine cells may communicate with exocrine cells directly through cytokines or indirectly by regulating the immune microenvironment. This crosstalk affects the onset and progression of these diseases. This review summarizes the history and milestones of findings related to the exocrine and endocrine pancreas, their embryonic development, phenotypic transformations, signaling roles in health and disease, the endocrine-exocrine crosstalk from the perspective of diseases, and potential therapeutic targets. Elucidating the regulatory mechanisms of pancreatic endocrine and exocrine signaling and provide novel insights for the understanding and treatment of diseases.
Collapse
Grants
- National High Level Hospital Clinical Research Funding (2022, 2022-PUMCH-D-001, to YZ), CAMS Innovation Fund for Medical Sciences (2021, 2021-I2M-1-002, to YZ), National Nature Science Foundation of China (2021, 82102810, to CW, the Fundamental Research Funds for the Central Universities(3332023123)
- cNational High Level Hospital Clinical Research Funding (2022, 2022-PUMCH-D-001, to YZ), CAMS Innovation Fund for Medical Sciences (2021, 2021-I2M-1-002, to YZ), National Nature Science Foundation of China (2021, 82102810, to CW, the Fundamental Research Funds for the Central Universities(3332023123)
Collapse
Affiliation(s)
- Chenglin Hu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China
| | - Yuan Chen
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China
| | - Xinpeng Yin
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China
| | - Ruiyuan Xu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China
| | - Chenxue Yin
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China
| | - Chengcheng Wang
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China.
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China.
- National Infrastructures for Translational Medicine, Peking Union Medical College Hospital, Beijing, PR China.
- Institute of Clinical Medicine, Peking Union Medical College Hospital, Beijing, PR China.
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China.
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China.
- National Infrastructures for Translational Medicine, Peking Union Medical College Hospital, Beijing, PR China.
| |
Collapse
|
10
|
Wu Y, Wang H, Xu H. Autophagy-lysosome pathway in insulin & glucagon homeostasis. Front Endocrinol (Lausanne) 2025; 16:1541794. [PMID: 39996055 PMCID: PMC11847700 DOI: 10.3389/fendo.2025.1541794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 01/22/2025] [Indexed: 02/26/2025] Open
Abstract
Lysosome, a highly dynamic organelle, is an important nutrient sensing center. They utilize different ion channels and transporters to complete the mission in degradation, trafficking, nutrient sensing and integration of various metabolic pathways to maintain cellular homeostasis. Glucose homeostasis relies on tightly regulated insulin secretion by pancreatic β cells, and their dysfunction is a hallmark of type 2 diabetes. Glucagon also plays an important role in hyperglycemia in diabetic patients. Currently, lysosome has been recognized as a nutrient hub to regulate the homeostasis of insulin and other hormones. In this review, we will discuss recent advances in understanding lysosome-mediated autophagy and lysosomal proteins involved in maintaining insulin and glucagon homeostasis, as well as their contributions to the etiology of diabetes.
Collapse
Affiliation(s)
- Yi Wu
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Shanghai Key Laboratory of Molecular Imaging, School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Hui Wang
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Huoyan Xu
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
| |
Collapse
|
11
|
Na YJ, Choi KJ, Jung WH, Park SB, Koh B, Hoe KL, Kim KY. Development of 3D Muscle Cell Culture-Based Screening System for Metabolic Syndrome Drug Research. Tissue Eng Part C Methods 2025; 31:53-64. [PMID: 39912898 DOI: 10.1089/ten.tec.2024.0292] [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: 02/07/2025] Open
Abstract
Developing effective drug screening methods for type 2 diabetes requires physiologically relevant models. Traditional 2D cell cultures have limitations in replicating in vivo conditions, leading to challenges in assessing drug efficacy. To overcome these issues, we developed a 3D artificial muscle model that induces insulin resistance, a hallmark of type 2 diabetes. Using C2C12 myoblasts cultured in a scaffold of 1% alginate and 1 mg/mL collagen type 1, we optimized conditions for differentiation and structural stability. Insulin resistance was induced using palmitic acid (PA), and glucose uptake was assessed using the fluorescent glucose analog 2-NBDG. The 3D model demonstrated superior glucose uptake responses compared with 2D cultures, with a threefold increase in insulin-stimulated glucose uptake on days 4 and 8 of differentiation. Induced insulin resistance was observed with 0.1 mM PA, which maintained cell viability and differentiation capacity. The model was validated through comparative drug screening using rosiglitazone and metformin, as well as 165 candidate compounds provided by Korea Chemical Bank. Drug screening revealed that three out of five hit compounds identified in both 2D and 3D models exhibited greater efficacy in 3D cultures, with results consistent with ex vivo assays using mouse soleus muscle. This model closely mimics in vivo conditions, offering a robust platform for type 2 diabetes drug discovery while supporting ethical research practices.
Collapse
Affiliation(s)
- Yoon-Ju Na
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, Korea
- Department of New Drug Discovery and Development, Chungnam National University, Daejeon, Korea
| | - Kyoung Jin Choi
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, Korea
| | - Won Hoon Jung
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, Korea
| | - Sung Bum Park
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, Korea
| | - Byumseok Koh
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, Korea
| | - Kwang-Lae Hoe
- Department of New Drug Discovery and Development, Chungnam National University, Daejeon, Korea
| | - Ki Young Kim
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, Korea
- Department of New Drug Discovery and Development, Chungnam National University, Daejeon, Korea
| |
Collapse
|
12
|
Rendell M. Pharmacotherapy of type 1 diabetes - part 1: yesterday. Expert Opin Pharmacother 2025; 26:313-324. [PMID: 39875200 DOI: 10.1080/14656566.2025.2454280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 01/13/2025] [Indexed: 01/30/2025]
Abstract
INTRODUCTION Type 1 diabetes is a unique autoimmune attack on the β cell of the pancreatic islet resulting in progressive destruction of these cells and as a result the ability of the body to maintain insulin production. The consequences of insulin deficiency are very severe, and the disease was fatal prior to the ability to extract insulin from animal pancreas in 1921. We review progress in the treatment of childhood type 1 diabetes over the past 100 years. AREAS COVERED We used PubMed and standard search engines to search for the evolution of diagnosis and treatment of type 1 diabetes. EXPERT OPINION Insulin replacement proved lifesaving for children afflicted with type 1 diabetes. However, it was observed that these children suffered from microvascular and large vessel disease. The Diabetes Control and Complications Trial (DCCT) with its extension Epidemiology of Diabetes Interventions and Complications Trial (EDIC) proved that control of blood glucose as close to normal as possible could prevent these diabetes-related conditions. Many formuations of insulin with varying onset and duration of action have been developed; yet normalization of glucose levels is difficult due to hypoglycemic events. There is continued progress toward that goal.
Collapse
Affiliation(s)
- Marc Rendell
- The Association of Diabetes Investigators, Newport Coast, CA, USA
- The Rose Salter Medical Research Foundation, Newport Coast, CA, USA
| |
Collapse
|
13
|
Ma H, Peng G, Hu Y, Lu B, Zheng Y, Wu Y, Feng W, Shi Y, Pan X, Song L, Stützer I, Liu Y, Fei J. Revealing the biological features of the axolotl pancreas as a new research model. Front Cell Dev Biol 2025; 13:1531903. [PMID: 39958891 PMCID: PMC11825805 DOI: 10.3389/fcell.2025.1531903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 01/07/2025] [Indexed: 02/18/2025] Open
Abstract
Introduction The pancreas plays a crucial role in digestion and blood glucose regulation. Current animal models, primarily mice and zebrafish, have limited the exploration of pancreatic biology from an evolutionary-developmental perspective. Tetrapod vertebrate axolotl (Ambystoma mexicanum) serves as a valuable model in developmental, regenerative, and evolutionary biology. However, the fundamental biology of the axolotl pancreas remains underexplored. This study aims to characterize the unique developmental, functional, and evolutionary features of the axolotl pancreas to expand the understanding of pancreatic biology. Methods We conducted morphological, histological, and transcriptomic analyses to investigate the axolotl pancreas. Pancreatic development was observed using in situ hybridization and immunostaining for key pancreatic markers. RNA sequencing was performed to profile global gene expression during larva and adult stages. And differential gene expression analysis was used to characterize the conserved and unique gene patterns in the axolotl pancreas. Functional assays, including glucose tolerance tests and insulin tolerance tests, were optimized for individual axolotls. To assess pancreatic gene function, Pdx1 mutants were generated using CRISPR/Cas9-mediated gene editing, and their effects on pancreatic morphology, endocrine cell populations, and glucose homeostasis were analyzed. Results The axolotl pancreas contains all known pancreatic cell types and develops from dorsal and ventral buds. Both of buds contribute to exocrine and endocrine glands. The dorsal bud produces the major endocrine cell types, while the ventral bud generates α and δ cells, but not β cells. Differential gene expression analysis indicated a transition in global gene expression from pancreatic cell fate commitment and the cell cycle to glucose response, hormone synthesis, and secretion, following the development progression. Notably, the adult axolotl pancreas exhibits slower metabolic activity compared to mammals, as evidenced by the results of GTT and ITT. The mutation of Pdx1 resulted in hyperglycemia and a significant reduction in pancreatic cell mass, including a complete loss of endocrine cells, although it did not lead to a lethal phenotype. Discussion This study examines the axolotl pancreas, highlighting the conservation of pancreatic development. Our study highlights the unique features of the axolotl pancreas and broadens the scope of animal models available for pancreatic evolution and disease research.
Collapse
Affiliation(s)
- Hui Ma
- Department of Pathology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
- BGI Research, Qingdao, China
- School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Guangcong Peng
- Key Laboratory of Brain, Cognition and Education Sciences, Guangdong Key Laboratory of Mental Health and Cognitive Science, Ministry of Education, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China
| | - Yan Hu
- Department of Pathology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Binbin Lu
- Department of Pathology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yiying Zheng
- Key Laboratory of Brain, Cognition and Education Sciences, Guangdong Key Laboratory of Mental Health and Cognitive Science, Ministry of Education, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China
| | - Yingxian Wu
- Key Laboratory of Brain, Cognition and Education Sciences, Guangdong Key Laboratory of Mental Health and Cognitive Science, Ministry of Education, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China
| | - Weimin Feng
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Guangdong Medical University, Dongguan, China
| | - Yu Shi
- Key Laboratory of Brain, Cognition and Education Sciences, Guangdong Key Laboratory of Mental Health and Cognitive Science, Ministry of Education, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China
| | - Xiangyu Pan
- Department of Pathology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Li Song
- Department of Pathology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Ina Stützer
- Deutsche Forschungsgemeinschaft (DFG)-Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Yanmei Liu
- Key Laboratory of Brain, Cognition and Education Sciences, Guangdong Key Laboratory of Mental Health and Cognitive Science, Ministry of Education, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China
| | - Jifeng Fei
- Department of Pathology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
- School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
14
|
Xiang Y, Zhang C, Wang J, Cheng Y, Wang K, Wang L, Tong Y, Yan D. Identification of Metabolic Characteristic-Pancreatic Ductal Adenocarcinoma Associations Using Mendelian Randomization and Metabolomics. J Gastrointest Cancer 2025; 56:48. [PMID: 39833419 PMCID: PMC11753325 DOI: 10.1007/s12029-025-01173-4] [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: 01/11/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Metabolic reprogramming is increasingly recognized as a crucial factor influencing the development, progression, and prognosis of pancreatic ductal adenocarcinoma (PDAC). Despite this, the potential association of specific metabolic characteristics and PDAC remains ambiguous due to the variability introduced by individual patient differences. In this study, we aimed to find out metabolic pathways that may be associated with the overall survival (OS) of PDAC patients. METHODS We utilized Mendelian randomization (MR) to assess the associations between 1400 metabolites and metabolite ratios and PDAC. We performed functional annotation and pathway enrichment analysis on both significant metabolites and the shared proteins corresponding to the significant metabolite ratios. Additionally, we analyzed peripheral blood metabolites from 32 PDAC patients to correlate metabolites with clinicopathological features and OS. Functional enrichment analysis was also conducted on the significant metabolites. RESULTS Our MR analysis revealed 55 metabolites/metabolite ratios associated with PDAC. Among the top 20 enriched metabolic pathways involving proteins related to significant metabolite ratios, seven were associated with amino acid metabolism, three with carbohydrate metabolism, and two with lipid metabolism. Serum metabolomics of PDAC patients highlighted significant upregulation in pathways related to primary bile acid biosynthesis, as well as taurine and hypotaurine metabolism, which correlated negatively with OS. Conversely, pathways involved in arginine biosynthesis, arginine and proline metabolism, and aminoacyl-tRNA biosynthesis were notably downregulated and positively associated with OS. Both upregulated and downregulated differential metabolites were notably enriched in the pyrimidine metabolism pathway, which was linked to poorer OS. These associations were corroborated by MR analysis. CONCLUSION The study provides valuable insights into the metabolic characteristics associated with PDAC, offering a reference point for improving diagnosis and treatment for PDAC.
Collapse
Affiliation(s)
- Yaoxian Xiang
- Department of Oncology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, 101149, China
| | - Chan Zhang
- Department of Oncology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, 101149, China
| | - Jing Wang
- Department of Oncology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, 101149, China
| | - Yurong Cheng
- Department of Oncology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, 101149, China
| | - Kangjie Wang
- Department of Oncology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, 101149, China
| | - Li Wang
- Department of Oncology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, 101149, China
| | - Yingying Tong
- Department of Oncology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, 101149, China
| | - Dong Yan
- Department of Oncology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, 101149, China.
| |
Collapse
|
15
|
Šestan M, Raposo B, Rendas M, Brea D, Pirzgalska R, Rasteiro A, Aliseychik M, Godinho I, Ribeiro H, Carvalho T, Wueest S, Konrad D, Veiga-Fernandes H. Neuronal-ILC2 interactions regulate pancreatic glucagon and glucose homeostasis. Science 2025; 387:eadi3624. [PMID: 39818880 DOI: 10.1126/science.adi3624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 04/02/2024] [Accepted: 11/08/2024] [Indexed: 01/19/2025]
Abstract
The immune system shapes body metabolism, while interactions between peripheral neurons and immune cells control tissue homeostasis and immunity. However, whether peripheral neuroimmune interactions orchestrate endocrine system functions remains unexplored. After fasting, mice lacking type 2 innate lymphoid cells (ILC2s) displayed disrupted glucose homeostasis, impaired pancreatic glucagon secretion, and inefficient hepatic gluconeogenesis. Additionally, intestinal ILC2s were found in the pancreas, which was dependent on their expression of the adrenergic beta 2 receptor. Targeted activation of catecholaminergic intestinal neurons promoted the accumulation of ILC2s in the pancreas. Our work provides evidence that immune cells can be regulated by neuronal signals in response to fasting, activating an inter-organ communication route that promotes pancreatic endocrine function and regulation of blood glucose levels.
Collapse
Affiliation(s)
- Marko Šestan
- Champalimaud Foundation. Champalimaud Centre for the Unknown. Champalimaud Research. Lisbon, Portugal
| | - Bruno Raposo
- Champalimaud Foundation. Champalimaud Centre for the Unknown. Champalimaud Research. Lisbon, Portugal
| | - Miguel Rendas
- Champalimaud Foundation. Champalimaud Centre for the Unknown. Champalimaud Research. Lisbon, Portugal
| | - David Brea
- Champalimaud Foundation. Champalimaud Centre for the Unknown. Champalimaud Research. Lisbon, Portugal
| | - Roksana Pirzgalska
- Champalimaud Foundation. Champalimaud Centre for the Unknown. Champalimaud Research. Lisbon, Portugal
| | - Ana Rasteiro
- Champalimaud Foundation. Champalimaud Centre for the Unknown. Champalimaud Research. Lisbon, Portugal
| | - Maria Aliseychik
- Champalimaud Foundation. Champalimaud Centre for the Unknown. Champalimaud Research. Lisbon, Portugal
| | - Inês Godinho
- Champalimaud Foundation. Champalimaud Centre for the Unknown. Champalimaud Research. Lisbon, Portugal
| | - Hélder Ribeiro
- Champalimaud Foundation. Champalimaud Centre for the Unknown. Champalimaud Research. Lisbon, Portugal
| | - Tania Carvalho
- Champalimaud Foundation. Champalimaud Centre for the Unknown. Champalimaud Research. Lisbon, Portugal
| | - Stephan Wueest
- Division of Pediatric Endocrinology and Diabetology, University Children's Hospital, University of Zurich, Zurich, Switzerland
- Children's Research Center, University Children's Hospital, University of Zurich, Zurich, Switzerland
| | - Daniel Konrad
- Division of Pediatric Endocrinology and Diabetology, University Children's Hospital, University of Zurich, Zurich, Switzerland
- Children's Research Center, University Children's Hospital, University of Zurich, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Henrique Veiga-Fernandes
- Champalimaud Foundation. Champalimaud Centre for the Unknown. Champalimaud Research. Lisbon, Portugal
| |
Collapse
|
16
|
Ge Y. Integrating New Approach Methodologies to Address Environmental Pancreatic Toxicity and Metabolic Disorders. BIOLOGY 2025; 14:85. [PMID: 39857315 PMCID: PMC11762660 DOI: 10.3390/biology14010085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/06/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
Advancing our understanding of pancreatic toxicity and metabolic disorders caused by environmental exposures requires innovative approaches. The pancreas, a vital organ for glucose regulation, is increasingly recognized as a target of harm from environmental chemicals and dietary factors. Traditional toxicological methods, while foundational, often fail to address the mechanistic complexities of pancreatic dysfunction, particularly under real-world conditions involving multiple exposures. New Approach Methodologies (NAMs)-including high-throughput screening (HTS), OMICS technologies, computational modeling, and advanced in vitro systems-offer transformative tools to tackle these challenges. NAMs enable the identification of mechanistic pathways, improve testing efficiency, and reduce reliance on animal testing. This commentary explores the integration of NAMs into pancreatic toxicity screening, addresses critical gaps in evaluating the cumulative risks of chemical and dietary exposures, and proposes solutions for integrating the pancreas into toxicity screening through NAMs. By highlighting recent advancements and emphasizing their adoption in environmental toxicity assessment frameworks, this work demonstrates the potential of NAMs to revolutionize environmental health research, inspire interdisciplinary collaboration, and protect public health.
Collapse
Affiliation(s)
- Yue Ge
- Center for Computational Toxicology and Exposure, US Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| |
Collapse
|
17
|
Ahmed S, Adnan H, Khawaja MA, Butler AE. Novel Micro-Ribonucleic Acid Biomarkers for Early Detection of Type 2 Diabetes Mellitus and Associated Complications-A Literature Review. Int J Mol Sci 2025; 26:753. [PMID: 39859467 PMCID: PMC11765584 DOI: 10.3390/ijms26020753] [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: 12/17/2024] [Revised: 01/11/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM) is one of the most widespread chronic diseases globally, with its prevalence expected to rise significantly in the years ahead. Previous studies on risk stratification for T2DM identify certain biomarkers, including glycated hemoglobin (HbA1c), oral glucose tolerance testing (OGTT), fructosamine, and glycated albumin, as key indicators for predicting the onset and progression of T2DM. However, these traditional markers have been shown to lack sensitivity and specificity and their results are difficult to analyze due to non-standardized interpretation criteria, posing significant challenges to an accurate and definitive diagnosis. The strict measures of these traditional markers may not catch gradual increases in blood sugar levels during the early stages of diabetes evolution, as these might still fall within acceptable glycemic parameters. Recent advancements in research have suggested novel micro ribonucleic acid (miRNA) as circulatory molecules that can facilitate the early detection of prediabetic conditions in high-risk groups and potentially enable prevention of the progression to T2DM. This capability makes them a very powerful tool for potentially improving population health, enhancing outcomes for many patients, and reducing the overall burden of T2DM. These promising biomarkers are small, noncoding RNA involved in the regulation of many cellular functions that have a hand in the metabolic activities of cells, making them a very useful and relevant biomarker to explore for the diagnosis and risk stratification of T2DM. This review analyzes the current literature, outlining the occurrence of miRNAs in prediabetic and diabetic individuals and their implications in predicting dysglycemic disorders.
Collapse
Affiliation(s)
- Sara Ahmed
- School of Medicine, Royal College of Surgeons in Ireland-Bahrain, Busaiteen 15503, Bahrain; (S.A.); (H.A.); (M.A.K.)
| | - Haroon Adnan
- School of Medicine, Royal College of Surgeons in Ireland-Bahrain, Busaiteen 15503, Bahrain; (S.A.); (H.A.); (M.A.K.)
| | - Maryam A. Khawaja
- School of Medicine, Royal College of Surgeons in Ireland-Bahrain, Busaiteen 15503, Bahrain; (S.A.); (H.A.); (M.A.K.)
| | - Alexandra E. Butler
- Research Department, Royal College of Surgeons in Ireland-Bahrain, Busaiteen 15503, Bahrain
| |
Collapse
|
18
|
Liu Y, Wang W, Liang B, Zou Z, Zhang A. NLRP3 inflammasome activation and disruption of IRS-1/PI3K/AKT signaling: Potential mechanisms of arsenic-induced pancreatic beta cells dysfunction in rats. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 289:117504. [PMID: 39657381 DOI: 10.1016/j.ecoenv.2024.117504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/27/2024] [Accepted: 12/06/2024] [Indexed: 12/12/2024]
Abstract
Environmental exposure to arsenic is associated with significant health risks, including diabetogenic effects linked to pancreatic dysfunction. The NOD-like receptor protein 3 (NLRP3) inflammasome has been implicated in various metabolic abnormalities; however, its specific role in arsenic-induced pancreatic dysfunction remains insufficiently understood. This study aimed to elucidate the involvement and underlying mechanisms of the NLRP3 inflammasome in arsenic-induced pancreatic beta cells dysfunction through in vivo and in vitro models. In rat models, arsenic exposure was found to activate the NLRP3 inflammasome, as evidenced by pathomorphological changes and the expression of inflammasome activation markers. These pathological changes were accompanied by disruptions in the insulin signaling pathway, characterized by increased phosphorylation of insulin receptor substrate 1 (IRS-1) at Ser616, reduced expression of phosphatidylinositol 3-kinase (PI3K) and phosphorylated protein kinase B (AKT) at Ser473, and significant decreases in downstream targets, including the nuclear translocation of PDX-1, membrane translocation of glucose transporter 2 (GLUT2), and glucokinase (GCK) expression. In vitro, NaAsO2-treated INS-1 cells exhibited a dose-dependent reduction in glucose-stimulated insulin secretion. Furthermore, arsenic exposure in these cells activated the NLRP3 inflammasome, suppressed the IRS-1/PI3K/AKT signaling pathway, and downregulated insulin secretion regulatory molecules (PDX-1, GLUT2, and GCK). Notably, these arsenic-induced effects were reversed by MCC950, an NLRP3 inflammasome inhibitor, and Extendin-4, an agonist of the IRS-1/PI3K/AKT signaling pathway. Collectively, these findings demonstrate that NLRP3 inflammasome activation disrupts the IRS-1/PI3K/AKT signaling pathway, contributing to arsenic-induced pancreatic beta cells dysfunction in rats.
Collapse
Affiliation(s)
- Yonglian Liu
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang, Guizhou 550025, PR China
| | - Wenjuan Wang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang, Guizhou 550025, PR China
| | - Bing Liang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang, Guizhou 550025, PR China
| | - Zhonglan Zou
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang, Guizhou 550025, PR China
| | - Aihua Zhang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang, Guizhou 550025, PR China.
| |
Collapse
|
19
|
Gottmann P, Speckmann T, Stadion M, Chawla P, Saurenbach J, Ninov N, Lickert H, Schürmann A. Transcriptomic heterogeneity of non-beta islet cells is associated with type 2 diabetes development in mouse models. Diabetologia 2025; 68:166-185. [PMID: 39508880 DOI: 10.1007/s00125-024-06301-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 09/04/2024] [Indexed: 11/15/2024]
Abstract
AIMS/HYPOTHESIS The aim of this work was to understand the role of non-beta cells in pancreatic islets at early stages of type 2 diabetes pathogenesis. METHODS Specific clustering was employed to single-cell transcriptome data from islet cells of obese mouse strains differing in their diabetes susceptibility (diabetes-resistant B6.V.Lepob/ob [OB] and diabetes-susceptible New Zealand Obese [NZO] mice) on a diabetogenic diet. RESULTS Refined clustering analysis revealed several heterogeneous subpopulations for alpha cells, delta cells and macrophages, of which 133 mapped to human diabetes genes identified by genome-wide association studies. Importantly, a similar non-beta cell heterogeneity was found in a dataset of human islets from donors at different stages of type 2 diabetes. The predominant alpha cell cluster in NZO mice displayed signs of cellular stress and lower mitochondrial capacity (97 differentially expressed genes [DEGs]), whereas delta cells from these mice exhibited higher expression levels of maturation marker genes (Hhex and Sst) but lower somatostatin secretion than OB mice (184 DEGs). Furthermore, a cluster of macrophages was almost twice as abundant in islets of OB mice, and displayed extensive cell-cell communication with beta cells of OB mice. Treatment of beta cells with IL-15, predicted to be released by macrophages, activated signal transducer and activator of transcription (STAT3), which may mediate anti-apoptotic effects. Similar to mice, humans without diabetes possess a greater number of macrophages than those with prediabetes (39 mmol/mol [5.7%] < HbA1c < 46 mmol/mol [6.4%]) and diabetes. CONCLUSIONS/INTERPRETATION Our study indicates that the transcriptional heterogeneity of non-beta cells has an impact on intra-islet crosstalk and participates in beta cell (dys)function. DATA AVAILABILITY scRNA-seq data from the previous study are available in gene expression omnibus under gene accession number GSE159211 ( https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE159211 ).
Collapse
Affiliation(s)
- Pascal Gottmann
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany
- German Center for Diabetes Research (DZD), München Neuherberg, Germany
| | - Thilo Speckmann
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany
- German Center for Diabetes Research (DZD), München Neuherberg, Germany
| | - Mandy Stadion
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany
- German Center for Diabetes Research (DZD), München Neuherberg, Germany
| | - Prateek Chawla
- German Center for Diabetes Research (DZD), München Neuherberg, Germany
- Center for Regenerative Therapies TU Dresden, Dresden, Germany
| | - Judith Saurenbach
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany
- German Center for Diabetes Research (DZD), München Neuherberg, Germany
| | - Nikolay Ninov
- German Center for Diabetes Research (DZD), München Neuherberg, Germany
- Center for Regenerative Therapies TU Dresden, Dresden, Germany
| | - Heiko Lickert
- German Center for Diabetes Research (DZD), München Neuherberg, Germany
- Institute of Diabetes and Regeneration Research, Helmholtz Center Munich, Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Center Munich, Neuherberg, Germany
| | - Annette Schürmann
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany.
- German Center for Diabetes Research (DZD), München Neuherberg, Germany.
- University of Potsdam, Institute of Nutritional Sciences, Nuthetal, Germany.
| |
Collapse
|
20
|
Chen W, Yu S, Webber B, DeWolf EL, Kilmer R, Xian S, Schmidt CR, Power EM, Webber MJ. Supramolecular Peptide Depots for Glucose-Responsive Glucagon Delivery. J Biomed Mater Res A 2025; 113:e37854. [PMID: 39719890 DOI: 10.1002/jbm.a.37854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/04/2024] [Accepted: 12/09/2024] [Indexed: 12/26/2024]
Abstract
Precise blood glucose control continues to be a critical challenge in the treatment and management of type 1 diabetes in order to mitigate both acute and chronic complications. This study investigates the development of a supramolecular peptide amphiphile (PA) material functionalized with phenylboronic acid (PBA) for glucose-responsive glucagon delivery. The PA-PBA system self-assembles into nanofibrillar hydrogels in the presence of physiological glucose levels, resulting in stable hydrogels capable of releasing glucagon under hypoglycemic conditions. Glucose responsiveness is driven by reversible binding between PBA and glucose, which modulates the electrostatic interactions necessary for hydrogel formation and dissolution. Through comprehensive in vitro characterization, including circular dichroism, zeta potential measurements, and rheological assessments, the PA-PBA system is found to exhibit glucose-dependent assembly, enabling controlled glucagon release that is inversely related to glucose concentration. Glucagon release is accelerated under low glucose conditions, simulating a hypoglycemic state, with a reduced rate seen at higher glucose levels. Evaluation of the platform in vivo using a type 1 diabetic mouse model demonstrates the efficacy in protecting against insulin-induced hypoglycemia by restoring blood glucose levels following an insulin overdose. The ability to tailor glucagon release in response to fluctuating glucose concentrations underscores the potential of this platform for improving glycemic control. These findings suggest that glucose-stabilized supramolecular peptide hydrogels hold significant promise for responsive drug delivery applications, offering an approach to manage glucose levels in diabetes and other metabolic disorders.
Collapse
Affiliation(s)
- Weike Chen
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, USA
| | - Sihan Yu
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, USA
| | - Bernice Webber
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, USA
| | - Emily L DeWolf
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, USA
| | - Rory Kilmer
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, USA
| | - Sijie Xian
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, USA
| | - Connor R Schmidt
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, USA
| | - Elizabeth M Power
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, USA
| | - Matthew J Webber
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, USA
| |
Collapse
|
21
|
Yazawa K, Nakashima M, Nakagawa T, Yanase Y, Yoda Y, Ozawa K, Hosoi T. Pancreatic β cell-secreted factor FGF23 attenuates Alzheimer's disease-related amyloid β-induced neuronal death. PNAS NEXUS 2025; 4:pgae542. [PMID: 39876880 PMCID: PMC11773612 DOI: 10.1093/pnasnexus/pgae542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 11/18/2024] [Indexed: 01/31/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by cognitive decline and memory impairment. The pathophysiology of AD may involve aggregated amyloid β (Aβ) accumulation, which may underlie the disease mechanism. Patients with diabetes exhibit an elevated risk of developing AD, indicating potential therapeutic implications upon elucidating the underlying mechanisms. We hypothesized that pancreatic β cell-secreted factors could protect neurons from Aβ-induced toxicity. Therefore, we established an experimental model to elucidate the communication between pancreatic β cells and neuronal cells. Notably, our findings demonstrate that pancreatic β cell culture supernatant effectively inhibits Aβ-induced neuronal cell death. Transcriptomic analysis showed significant up-regulation of multiple ribosomal protein genes in neuronal cells treated with pancreatic β cell culture supernatant. Fibroblast growth factor 23, a secreted factor from pancreatic β cells, significantly suppressed Aβ-induced neuronal cell death. Our findings suggest that pancreatic β cells may secrete previously unrecognized neuroprotective factors, thereby attenuating neuronal cell death in AD.
Collapse
Affiliation(s)
- Kyosuke Yazawa
- Department of Pharmacotherapy, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Mieko Nakashima
- Department of Pharmacotherapy, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Tadashi Nakagawa
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, 1-1-1 Daigaku-dori, Sanyo Onoda City, Yamaguchi 756-0884, Japan
| | - Yuhki Yanase
- Department of Pharmacotherapy, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Yukari Yoda
- Department of Pharmacotherapy, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Koichiro Ozawa
- Department of Pharmacotherapy, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Toru Hosoi
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, 1-1-1 Daigaku-dori, Sanyo Onoda City, Yamaguchi 756-0884, Japan
| |
Collapse
|
22
|
Cichosz SL, Kronborg T, Laugesen E, Hangaard S, Fleischer J, Hansen TK, Jensen MH, Poulsen PL, Vestergaard P. From Stability to Variability: Classification of Healthy Individuals, Prediabetes, and Type 2 Diabetes Using Glycemic Variability Indices from Continuous Glucose Monitoring Data. Diabetes Technol Ther 2025; 27:34-44. [PMID: 39115921 DOI: 10.1089/dia.2024.0226] [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] [Indexed: 08/28/2024]
Abstract
Objective: This study aims to investigate the continuum of glucose control from normoglycemia to dysglycemia (HbA1c ≥ 5.7%/39 mmol/mol) using metrics derived from continuous glucose monitoring (CGM). In addition, we aim to develop a machine learning-based classification model to classify dysglycemia based on observed patterns. Methods: Data from five distinct studies, each featuring at least two days of CGM, were pooled. Participants included individuals classified as healthy, with prediabetes, or with type 2 diabetes mellitus (T2DM). Various CGM indices were extracted and compared across groups. The data set was split 70/30 for training and testing two classification models (XGBoost/Logistic Regression) to differentiate between prediabetes or dysglycemia and the healthy group. Results: The analysis included 836 participants (healthy: n = 282; prediabetes: n = 133; T2DM: n = 432). Across all CGM indices, a progressive shift was observed from the healthy group to those with diabetes (P < 0.001). Statistically significant differences (P < 0.01) were noted in mean glucose, time below range, time above 140 mg/dl, mobility, multiscale complexity index, and glycemic risk index when transitioning from health to prediabetes. The XGBoost models achieved the highest receiver operating characteristic area under the curve values on the test data set ranging from 0.91 [confidence interval (CI): 0.87-0.95] (prediabetes identification) to 0.97 [CI: 0.95-0.98] (dysglycemia identification). Conclusion: Our findings demonstrate a gradual deterioration of glucose homeostasis and increased glycemic variability across the spectrum from normo- to dysglycemia, as evidenced by CGM metrics. The performance of CGM-based indices in classifying healthy individuals and those with prediabetes and diabetes is promising.
Collapse
Affiliation(s)
- Simon Lebech Cichosz
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Thomas Kronborg
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
- Steno Diabetes Center North Denmark, Aalborg University Hospital, Aalborg, Denmark
| | - Esben Laugesen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Diagnostic Center, Regional Hospital Silkeborg, Silkeborg, Denmark
| | - Stine Hangaard
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
- Steno Diabetes Center North Denmark, Aalborg University Hospital, Aalborg, Denmark
| | - Jesper Fleischer
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Steno Diabetes Center Zealand, Zealand, Denmark
| | | | - Morten Hasselstrøm Jensen
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
- Department of Data Orchestration, Novo Nordisk, Søborg, Denmark
| | | | - Peter Vestergaard
- Steno Diabetes Center North Denmark, Aalborg University Hospital, Aalborg, Denmark
- Department of Endocrinology, Aalborg University Hospital, Aalborg, Denmark
| |
Collapse
|
23
|
Wang K, Zhang L, Deng B, Zhao K, Chen C, Wang W. Mitochondrial uncoupling protein 2: a central player in pancreatic disease pathophysiology. Mol Med 2024; 30:259. [PMID: 39707176 DOI: 10.1186/s10020-024-01027-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/03/2024] [Indexed: 12/23/2024] Open
Abstract
Pancreatic diseases pose considerable health challenges due to their complex etiology and limited therapeutic options. Mitochondrial uncoupling protein 2 (UCP2), highly expressed in pancreatic tissue, participates in numerous physiological processes and signaling pathways, indicating its potential relevance in these diseases. Despite this, UCP2's role in acute pancreatitis (AP) remains underexplored, and its functions in chronic pancreatitis (CP) and pancreatic steatosis are largely unknown. Additionally, the mechanisms connecting various pancreatic diseases are intricate and not yet fully elucidated. Given UCP2's diverse functionality, broad expression in pancreatic tissue, and the distinct pathophysiological features of pancreatic diseases, this review offers a comprehensive analysis of current findings on UCP2's involvement in these conditions. We discuss recent insights into UCP2's complex regulatory mechanisms, propose that UCP2 may serve as a central regulatory factor in pancreatic disease progression, and hypothesize that UCP2 dysfunction could significantly contribute to disease pathogenesis. Understanding UCP2's role and mechanisms in pancreatic diseases may pave the way for innovative therapeutic and diagnostic approaches.
Collapse
Affiliation(s)
- Kunpeng Wang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- General Surgery Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lilong Zhang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- General Surgery Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Beiying Deng
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Kailiang Zhao
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- General Surgery Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chen Chen
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China.
- General Surgery Laboratory, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Weixing Wang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China.
- General Surgery Laboratory, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
24
|
Lyu H, Kong J, Chen J, Zhang R, Xiao S, Guo D, Zhang Q, Chen XZ, Tang J, Zhou C. The Emerging Scenario of Ferroptosis in Pancreatic Cancer Tumorigenesis and Treatment. Int J Mol Sci 2024; 25:13334. [PMID: 39769097 PMCID: PMC11727763 DOI: 10.3390/ijms252413334] [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: 11/25/2024] [Revised: 12/10/2024] [Accepted: 12/10/2024] [Indexed: 01/05/2025] Open
Abstract
Pancreatic cancer remains one of the most lethal forms of cancer. Currently, there is a lack of effective drug treatments for pancreatic cancer. However, as a newly discovered form of non-apoptotic cell death, ferroptosis has garnered increasing attention in relation to pancreatic cancer. Understanding the role of ferroptosis in the tumorigenesis and treatment of pancreatic cancer may enable more effective clinical trials and treatments for pancreatic cancer and may minimize side effects or restrict the emergence of drug resistance. In this review, we summarize the current knowledge regarding the process and underlying mechanisms of ferroptosis, as well as its dual role in both promoting tumorigenesis and facilitating treatment strategies for pancreatic cancer. Additionally, how ferroptosis is implicated in the development of pancreatitis and insulin resistance, indicating that ferroptosis may play an important role in the risk of pancreatitis- and insulin-resistance-related pancreatic cancers, is also addressed.
Collapse
Affiliation(s)
- Hao Lyu
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Jinghua Kong
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Jiasi Chen
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Rui Zhang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Shuai Xiao
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Dong Guo
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Qi Zhang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Xing-Zhen Chen
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Jingfeng Tang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Cefan Zhou
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| |
Collapse
|
25
|
Wang S, Cheng X, Liang Z, Chen Z, Zhang J, Xu Q. Nocturnal Light Pollution Synergistically Impairs Glucose Metabolism With Age and Weight in Monkeys. J Diabetes Res 2024; 2024:5112055. [PMID: 39950097 PMCID: PMC11824604 DOI: 10.1155/2024/5112055] [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: 08/16/2024] [Accepted: 10/14/2024] [Indexed: 02/16/2025] Open
Abstract
Over the past decades, the global prevalence of Type 2 diabetes mellitus (T2D) and impaired glucose tolerance (IGT) has been increasing at an epidemic rate, yet the exact cause remains unknown. It is widely accepted that glucose metabolism can be impaired by circadian rhythms and sleep disturbances. Concurrently, exposures to light at night have been closely linked to circadian and sleep disturbances. However, there is no direct experiment on primates to demonstrate the precise extent of how serious light pollution impairs glucose metabolism, whether people will eventually become accustomed to this environment, and whether the pollution has synergistic impairing effects with aging and weight on glucose metabolism. To quantitatively address these questions, 137 cynomolgus were exposed to three distinct nocturnal light intensities for consecutive 10 months. Monthly glucose metabolism assessments were conducted. Data pertaining to the mortality rate of preexisting diabetes, incidence of light-induced diabetes and IGT, and alterations in insulin secretion were collected and analyzed. The results show that nocturnal light (1) caused premature deaths in individuals with preexisting diabetes; (2) intensity-dependently induced diabetes and IGT in previous healthy monkeys; (3) intensity-dependently reduced melatonin secretion; (4) had a synergistic impairing effect on glucose metabolism with aging and weight; and (5) although monkeys would eventually adapt to the environment, the disrupted glucose metabolism would not fully recover in most individuals. In conclusion, nocturnal light is associated with the global high prevalence of T2D and IGT. The harmful effects of light pollution on glucose metabolism are synergistic with age and weight.
Collapse
Affiliation(s)
- Shuxing Wang
- Department of Anatomy, Medical School, Foshan University, Foshan, Guangdong Province, China
| | - Xuange Cheng
- Department of Food and Pharmaceutical Sciences, Qingyuan Polytechnic, Panlong Park, Qingcheng District, Qingyuan City 511510, Guangdong Province, China
| | - Zihao Liang
- Qingyuan Hospital of Traditional Chinese Medicine, Qingyuan City, China
| | - Zhenyi Chen
- Primate Research Center, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, China
| | - Jiankai Zhang
- Department of Anatomy, Guangdong University, Dongguan, China
| | - Qiang Xu
- Primate Research Center, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, China
| |
Collapse
|
26
|
Ma J, Li X, Wang C. The Application of Nanomaterials in the Treatment of Pancreatic-Related Diseases. Int J Mol Sci 2024; 25:13158. [PMID: 39684868 DOI: 10.3390/ijms252313158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 11/16/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Pancreatic diseases, typically including pancreatic cancer, pancreatitis, and diabetes, pose enormous threats to people's lives and health. To date, therapeutics with high therapeutic efficacy and low side effects are still challenging. With the development of nanotechnology, nanomaterials have successfully been applied in pancretic disease treatment. Here, we first introduce the diversity of nanomaterials and the effects of their different physicochemical properties on pancreatic function. Following this, we analyze the potential of nanomaterials to enhance pancreatic targeting by overcoming the challenges of traditional delivery methods through surface modifications, structural adjustments, and optimized drug loading. Then, we introduce the application of structurally optimized nanomaterials to pancreatic-related diseases. For instance, on pancreatic cancer (as drug delivery platforms, for the promotion of radiation therapy, and as multifunctional tools), pancreatitis (as drug delivery systems, anti-inflammatory and anti-fibrotic agents), and diabetes (as insulin delivery carriers, for protecting pancreatic β cells, and for improving insulin resistance). Through analysis of the progress of current research, we summarize how nanomaterials can enhance treatment efficacy while minimizing side effects. Finally, we look forward to the prospects of nanomaterials in pancreatic disease treatment.
Collapse
Affiliation(s)
- Jing Ma
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xue Li
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunru Wang
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
27
|
Leontino G, Duval M. Homeostasis: understanding the effects of impaired mechanisms. BRITISH JOURNAL OF NURSING (MARK ALLEN PUBLISHING) 2024; 33:1094-1104. [PMID: 39639697 DOI: 10.12968/bjon.2024.0071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
This article focuses on homeostasis and offers a pathophysiological perspective. The dynamic mechanisms responsible for maintaining internal balance and disruptive processes will be analysed through the lens of key systems including the nervous, endocrine and renal systems. The environmental factors and their potential impact on homeostasis have been considered. A clinical case study will contextualise homeostasis in clinical practice.
Collapse
Affiliation(s)
- Giuseppe Leontino
- Senior Lecturer in Simulation and Clinical Skills, University of Hertfordshire
| | - Melanconia Duval
- Lecturer Practitioner in Critical care, University of West London
| |
Collapse
|
28
|
Pan C, Yu T, Zhao H, He J, Lu X, Tang H, Hong Y, Shang C, Wu Q, Yang A, Li C, Zhou M, Shi Y. Evaluation of pancreatic iodine uptake and related influential factors in multiphase dual-energy CT. Eur Radiol 2024; 34:7609-7621. [PMID: 38913243 DOI: 10.1007/s00330-024-10850-0] [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: 12/04/2023] [Revised: 04/25/2024] [Accepted: 06/03/2024] [Indexed: 06/25/2024]
Abstract
OBJECTIVES To establish normative values and identify potential factors influencing pancreatic iodine uptake using dual-energy CT (DECT). MATERIALS AND METHODS This retrospective study included participants without pancreatic diseases undergoing DECT at two institutions with different platforms. Their protocols both included arterial phase (AP), portal venous phase (PP), and equilibrium phase (EP), defined as 35 s-40 s, 60 s-70 s, and 150 s-180 s after injection of contrast agent, respectively. Both iodine concentration (IC) and normalised IC (NIC) were measured. Demographic features, local measurements of the pancreas and visceral fat area (VFA) were considered as potential factors influencing iodine uptake using multivariate linear regression analyses. RESULTS A total of 562 participants (median age 58 years [interquartile range: 47-67], with 282 men) were evaluated. The mean IC differed significantly between two institutions (all p < 0.001) across three contrast-enhanced phases, while the mean NIC showed no significant differences (all p > 0.05). The mean values of NIC were 0.22 at AP, 0.43 at PP and 0.45 at EP. NICAP was independently affected by VFA (β = 0.362, p < 0.001), smoking (β = -0.240, p = 0.001), and type-II diabetes (β = -0.449, p < 0.001); NICPP by VFA (β = -0.301, p = 0.017) and smoking (β = -0.291, p < 0.001); and NICEP by smoking (β = -0.154, p = 0.10) and alcohol consumption (β = -0.350, p < 0.001) with statistical power values over 0.81. CONCLUSION NIC values were consistent across institutions. Abdominal obesity, smoking, alcohol consumption, and diabetes are independent factors influencing pancreatic iodine uptake. CLINICAL RELEVANCE STATEMENT This study has provided reference normative values, influential factors and effective normalisation methods of pancreatic iodine uptake in multiphase dual-energy CT for future studies in this area as a new biological marker. KEY POINTS Evaluation of pancreatic iodine uptake measured by dual-energy CT is a promising method for future studies. Abdominal obesity, smoking, alcohol consumption, diabetes, and sex are independent factors influencing pancreatic iodine uptake. Utility of normalised iodine concentration is necessary to ensure the consistency across different institutions.
Collapse
Affiliation(s)
- Chen Pan
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Tao Yu
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Heng Zhao
- Department of Radiology, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Jiani He
- Department of Radiology, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Xiaomei Lu
- CT Clinical Science CT, Philips Healthcare, Shenyang, China
| | - Haiyan Tang
- Department of Radiology, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Yang Hong
- Department of Neurosurgery, Shengjing Hospital, China Medical University, Shenyang, China
| | - Chao Shang
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, China
| | - Qijun Wu
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Aoran Yang
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Chunli Li
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Minghui Zhou
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Yu Shi
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
29
|
Shezi M, Snyman C, Niesler CU. Candidate Gene Expression in Adult Zebrafish Models of Type 2 Diabetes Mellitus. Zebrafish 2024; 21:401-408. [PMID: 39527263 DOI: 10.1089/zeb.2024.0154] [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: 11/16/2024] Open
Abstract
Animal models are an important tool for studying noncommunicable diseases (NCDs) as they provide a unique opportunity to investigate real-time changes that occur in the onset of, and during, the diseased state. This is of particular importance given that the global prevalence of NCDs, such as type 2 diabetes mellitus (T2DM), is rising at an alarming rate. In South Africa, which has one of the highest levels of HIV in the world, the incidence of T2DM is thought to be associated, in part, with exposure to combination antiretrovirals. We report on the establishment of both nonobese and obese zebrafish models of T2DM, as well as associated changes in mRNA expression of preproinsulin and phosphoenolpyruvate carboxykinase (pck) 1 and 2. The diabetic state was achieved by either immersing adult zebrafish in a 2% glucose solution for 40 days or by overfeeding adult zebrafish for 10 weeks. Glucose immersion resulted in significantly elevated fasting blood glucose levels twice as high as control, whereas bodyweight did not change significantly (nonobese model). Overfeeding led to both significantly elevated fasting blood glucose and bodyweight compared with control (obese model). Both models were characterized by significantly increased preproinsulin mRNA expression indicating insulin resistance; mRNA expression of metabolic enzymes PCK 1 and 2 was also significantly upregulated, as seen in diabetic patients. These candidate gene expression changes, similar in both zebrafish models, establish a baseline that can be utilized to investigate the underlying mechanisms driving the increased T2DM incidence, using an excellent alternative to traditional rodent models.
Collapse
Affiliation(s)
- Mlondi Shezi
- Discipline of Biochemistry, School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Pietermaritzburg, South Africa
| | - Celia Snyman
- Discipline of Biochemistry, School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Pietermaritzburg, South Africa
| | - Carola Ulrike Niesler
- Discipline of Biochemistry, School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Pietermaritzburg, South Africa
| |
Collapse
|
30
|
Bisht S, Singh MF. The triggering pathway, the metabolic amplifying pathway, and cellular transduction in regulation of glucose-dependent biphasic insulin secretion. Arch Physiol Biochem 2024; 130:854-865. [PMID: 38196246 DOI: 10.1080/13813455.2023.2299920] [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: 08/07/2023] [Revised: 12/08/2023] [Accepted: 12/16/2023] [Indexed: 01/11/2024]
Abstract
INTRODUCTION Insulin secretion is a highly regulated process critical for maintaining glucose homeostasis. This abstract explores the intricate interplay between three essential pathways: The Triggering Pathway, The Metabolic Amplifying Pathway, and Cellular Transduction, in orchestrating glucose-dependent biphasic insulin secretion. MECHANISM During the triggering pathway, glucose metabolism in pancreatic beta-cells leads to ATP production, closing ATP-sensitive potassium channels and initiating insulin exocytosis. The metabolic amplifying pathway enhances insulin secretion via key metabolites like NADH and glutamate, enhancing calcium influx and insulin granule exocytosis. Additionally, the cellular transduction pathway involves G-protein coupled receptors and cyclic AMP, modulating insulin secretion. RESULT AND CONCLUSION These interconnected pathways ensure a dynamic insulin response to fluctuating glucose levels, with the initial rapid phase and the subsequent sustained phase. Understanding these pathways' complexities provides crucial insights into insulin dysregulation in diabetes and highlights potential therapeutic targets to restore glucose-dependent insulin secretion.
Collapse
Affiliation(s)
- Shradha Bisht
- Amity Institute of Pharmacy, Amity University, Lucknow, Uttar Pradesh, India
| | - Mamta F Singh
- School of Pharmaceutical Sciences, SBS University, Balawala, Uttarakhand, India
| |
Collapse
|
31
|
Vinciguerra D, Rajalakshmi P S, Yang J, Georgiou PG, Snell K, Pesenti T, Collins J, Tamboline M, Xu S, van Dam RM, Messina KMM, Hevener AL, Maynard HD. A Glucose-Responsive Glucagon-Micelle for the Prevention of Hypoglycemia. ACS CENTRAL SCIENCE 2024; 10:2036-2047. [PMID: 39634211 PMCID: PMC11613347 DOI: 10.1021/acscentsci.4c00937] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 12/07/2024]
Abstract
While glucose-responsive insulin delivery systems are in widespread clinical use to treat insulin insufficiency, the on-demand supplementation of glucagon for acute hypoglycemia treatment remains understudied. A self-regulated glucagon release material is highly desired to mitigate the potential risks of severe insulin-induced hypoglycemia. Here, we describe a glucose-responsive polymeric nanosystem with glucagon covalently grafted to the end-group. Under normoglycemic conditions, phenylboronic acid units in the polymer chain reversibly bind glucose, triggering self-assembly of the conjugate into micelles. During hypoglycemia, however, the micelle disassembles into its original, unimeric state, revealing the active glucagon conjugate. The formulation showed a 5-fold increase in activity compared to native glucagon when tested in vitro. Glucagon-loaded micelles injected into mice prevented or reversed deep hypoglycemia when administered prior to or during an insulin challenge. Glucagon release was only observed at or below the counterregulatory threshold and not during normoglycemia or moderate hypoglycemia. The in vivo acute and chronic toxicity analysis, along with μPET/μCT imaging, established the biosafety profile of this formulation and demonstrated no organ accumulation. This proof-of-concept work is the first step toward development of a translational, stimuli-responsive glucagon delivery platform to control glycemia.
Collapse
Affiliation(s)
- Daniele Vinciguerra
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
- California
NanoSystems Institute, University of California,
Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1569, United States
| | - Rajalakshmi P S
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
- California
NanoSystems Institute, University of California,
Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1569, United States
| | - Jane Yang
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
- California
NanoSystems Institute, University of California,
Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1569, United States
| | - Panagiotis G. Georgiou
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
- California
NanoSystems Institute, University of California,
Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1569, United States
| | - Katherine Snell
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
| | - Théo Pesenti
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
| | - Jeffrey Collins
- Department
of Molecular and Medical Pharmacology and Crump Institute for Molecular
Imaging, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095-1735, United States
| | - Mikayla Tamboline
- Department
of Molecular and Medical Pharmacology and Crump Institute for Molecular
Imaging, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095-1735, United States
| | - Shili Xu
- Department
of Molecular and Medical Pharmacology and Crump Institute for Molecular
Imaging, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095-1735, United States
- Jonsson
Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095-1735, United
States
| | - R. Michael van Dam
- Department
of Molecular and Medical Pharmacology and Crump Institute for Molecular
Imaging, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095-1735, United States
- Jonsson
Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095-1735, United
States
| | - Kathryn M. M. Messina
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
- California
NanoSystems Institute, University of California,
Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1569, United States
| | - Andrea L. Hevener
- Department
of Medicine, Division of Endocrinology, David Geffen School of Medicine at UCLA, 650 Charles E. Young Dr, Los Angeles, California 90095, United States
- VA
Greater
Los Angeles Healthcare System GRECC, Los Angeles, California 90073, United States
| | - Heather D. Maynard
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
- California
NanoSystems Institute, University of California,
Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1569, United States
| |
Collapse
|
32
|
Degen I, Robson Brown K, Reeve HWJ, Abdallah ZS. Beyond Expected Patterns in Insulin Needs of People With Type 1 Diabetes: Temporal Analysis of Automated Insulin Delivery Data. JMIRX MED 2024; 5:e44384. [PMID: 39654139 PMCID: PMC11612581 DOI: 10.2196/44384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 09/12/2024] [Accepted: 09/16/2024] [Indexed: 12/13/2024]
Abstract
Background Type 1 diabetes (T1D) is a chronic condition in which the body produces too little insulin, a hormone needed to regulate blood glucose. Various factors such as carbohydrates, exercise, and hormones impact insulin needs. Beyond carbohydrates, most factors remain underexplored. Regulating insulin is a complex control task that can go wrong and cause blood glucose levels to fall outside a range that protects people from adverse health effects. Automated insulin delivery (AID) has been shown to maintain blood glucose levels within a narrow range. Beyond clinical outcomes, data from AID systems are little researched; such systems can provide data-driven insights to improve the understanding and treatment of T1D. Objective The aim is to discover unexpected temporal patterns in insulin needs and to analyze how frequently these occur. Unexpected patterns are situations where increased insulin does not result in lower glucose or where increased carbohydrate intake does not raise glucose levels. Such situations suggest that factors beyond carbohydrates influence insulin needs. Methods We analyzed time series data on insulin on board (IOB), carbohydrates on board (COB), and interstitial glucose (IG) from 29 participants using the OpenAPS AID system. Pattern frequency in hours, days (grouped via k-means clustering), weekdays, and months were determined by comparing the 95% CI of the mean differences between temporal units. Associations between pattern frequency and demographic variables were examined. Significant differences in IOB, COB, and IG across temporal dichotomies were assessed using Mann-Whitney U tests. Effect sizes and Euclidean distances between variables were calculated. Finally, the forecastability of IOB, COB, and IG for the clustered days was analyzed using Granger causality. Results On average, 13.5 participants had unexpected patterns and 9.9 had expected patterns. The patterns were more pronounced (d>0.94) when comparing hours of the day and similar days than when comparing days of the week or months (0.3 Conclusions Our study shows that unexpected patterns in the insulin needs of people with T1D are as common as expected patterns. Unexpected patterns cannot be explained by carbohydrates alone. Our results highlight the complexity of glucose regulation and emphasize the need for personalized treatment approaches. Further research is needed to identify and quantify the factors that cause these patterns.
Collapse
Affiliation(s)
- Isabella Degen
- Interactive Artificial Intelligence Centre for Doctoral Training, School of Computer Science, Faculty of Science and Engineering, University of Bristol, 1 Cathedral Square, College Green, Bristol, BS1 5DD, United Kingdom, 44 7726100905
| | - Kate Robson Brown
- University College Dublin President's Office, College of Engineering and Architecture, University College Dublin, Dublin, Ireland
| | - Henry W J Reeve
- School of Mathematics, Faculty of Science and Engineering, University of Bristol, Bristol, United Kingdom
| | - Zahraa S Abdallah
- School of Engineering Mathematics and Technology, Faculty of Science and Engineering, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
33
|
Chen H, Mishra NK, Martos-Maldonado MC, Roholm S, Sørensen KK, Jensen KJ. Chemical Modification of Insulin Using Flow Chemistry. Chembiochem 2024; 25:e202400534. [PMID: 39166477 DOI: 10.1002/cbic.202400534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 08/23/2024]
Abstract
Chemical modification of proteins is of growing importance to generate new molecular probes for chemical biology and for the development of new biopharmaceuticals. For example, two approved, long-acting insulin variants are lipidated at the LysB29 side-chain. Acylations of proteins have so far been performed in batch-mode. Here we describe the use of flow chemistry for site-selective acylation of a small protein, insulin. To the best of our knowledge this is the first report on flow chemistry for chemical modification of insulin. The first step was to develop reaction conditions for acylation of Lys B29 that gave a soluble mixture and thus was compatible with flow chemistry in a microreactor; this included selection of a soluble base. Secondly, the conditions, such as reagent ratios and flow rate were optimized. Third, the use of these conditions for the acylation with a wide range of acids was demonstrated. Finally, Boc-protected insulins were synthesized. Insulin remained stable towards these flow chemistry conditions. This use of flow chemistry for the chemical modification of insulin opens the prospect of producing chemically modified biopharmaceuticals by flow chemistry with fewer byproducts.
Collapse
Affiliation(s)
- Haoyu Chen
- Department of Chemistry, University of Copenhgen, Thorvaldsensvej 40, DK-1871, Frederiksberg, Denmark
- Current address: Faculty for Chemistry and Pharmacy, Ludwig-Maximilian University, Butenandtstr 5-13, 81377, Munich, Germany
| | - Narendra Kumar Mishra
- Department of Chemistry, University of Copenhgen, Thorvaldsensvej 40, DK-1871, Frederiksberg, Denmark
| | - Manuel C Martos-Maldonado
- Department of Chemistry, University of Copenhgen, Thorvaldsensvej 40, DK-1871, Frederiksberg, Denmark
| | - Sandie Roholm
- Department of Chemistry, University of Copenhgen, Thorvaldsensvej 40, DK-1871, Frederiksberg, Denmark
| | - Kasper K Sørensen
- Department of Chemistry, University of Copenhgen, Thorvaldsensvej 40, DK-1871, Frederiksberg, Denmark
| | - Knud J Jensen
- Department of Chemistry, University of Copenhgen, Thorvaldsensvej 40, DK-1871, Frederiksberg, Denmark
| |
Collapse
|
34
|
Hwekwete A, Baiocchi C. Exploring homeostasis in children, the body's dynamic balancing act. Nurs Child Young People 2024:e1536. [PMID: 39552173 DOI: 10.7748/ncyp.2024.e1536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2024] [Indexed: 11/19/2024]
Abstract
Homeostasis can be described as the dynamic process within an organism designed to maintain a relatively constant internal environment that is conducive to the optimal functioning of cells, enzymes and organs. To regulate physiological variables, homeostasis relies on mechanisms called negative and positive feedback loops. However, certain factors can disrupt homeostatic processes, leading to dysregulation and disease. This article provides an overview of homeostasis, including negative and positive feedback loops and dysregulation. The authors use a fictional case study - an adolescent girl with type 1 diabetes who develops diabetic ketoacidosis - to illustrate the adverse effects of homeostatic dysregulation and demonstrate the relevance of understanding homeostasis in children's nursing.
Collapse
Affiliation(s)
- Agnes Hwekwete
- School of Life and Health Sciences, University of Roehampton, London, England
| | - Chiara Baiocchi
- College of Nursing, Midwifery and Healthcare, University of West London, London, England
| |
Collapse
|
35
|
Tagliasco M, Capuano E, Dall'Asta M, Renzetti S, Fogliano V, Pellegrini N. The combined effect of gluten addition and semolina cell wall integrity reduces the oral sugar release and the insulinemic response to bread in healthy volunteers. Eur J Nutr 2024; 64:6. [PMID: 39546030 DOI: 10.1007/s00394-024-03517-5] [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: 04/24/2024] [Accepted: 10/06/2024] [Indexed: 11/17/2024]
Abstract
PURPOSE A strategy to reduce starch digestibility is to limit its accessibility to α-amylase by preserving the integrity of cells where starch is encapsulated. Coarse flour is rich in intact cells and can be used for this purpose. However, making bread with coarse flour negatively affects crumb cohesiveness, which may increase the gastric disintegration rate, and enhance starch accessibility. Therefore, this study aimed to assess the combined effect of coarse semolina and its 20% gluten substitution in bread in healthy volunteers on glycemic and insulinemic responses, oral processing and bolus characteristics. METHODS Apparently, healthy volunteers (n = 16) randomly consumed bread made with coarse semolina and 20% gluten substitution (80CS_20G), its counterpart with fine semolina (80FS_20G), and bread with fine semolina and 5% gluten (95FS_5G). The glycemic and insulinemic responses were measured over 2 h after bread consumption. Mastication behaviour, bolus properties and reducing sugar were also evaluated. RESULTS No differences in glycemic responses and mastication were observed among the samples. 80CS_20G and 80FS_20G exhibited similar textural properties but 80CS_20G released less reducing sugars and elicited a lower insulin response at 30 min than 80FS_20G, probably due to intact cells that limit starch accessibility. Also, 95FS_5G released lower reducing sugars and had lower insulin peak than 80FS_20G. The compact structure of 95FS_5G may have delayed starch hydrolysis by restricting α-amylase accessibility. CONCLUSION Combining gluten and coarse semolina resulted in bread with a lower release of reducing sugars, a reduced insulinemic peak and textural properties similar to the counterpart with fine semolina. CLINICAL TRIAL REGISTRATION NUMBER The trial is registered at ClinicalTrials.gov: NCT06152874.
Collapse
Affiliation(s)
- Marianna Tagliasco
- Department of Agricultural, Food, Environmental and Animal Sciences, University of Udine, Via Sondrio 2/A, Udine, 33100, Italy
| | - Edoardo Capuano
- Food Quality and Design Group, Wageningen University, Bornse Weilanden 9, Wageningen, 6708 WG, The Netherlands
| | - Margherita Dall'Asta
- Department of Animal Science, Food and Nutrition, Università Cattolica del Sacro Cuore, Via Parmense 84, Piacenza, 29122, Italy
| | - Stefano Renzetti
- Wageningen Food and Biobased Research, Wageningen University & Research, Bornse Weilanden 9, Wageningen, 6708 WG, The Netherlands
| | - Vincenzo Fogliano
- Food Quality and Design Group, Wageningen University, Bornse Weilanden 9, Wageningen, 6708 WG, The Netherlands
| | - Nicoletta Pellegrini
- Department of Agricultural, Food, Environmental and Animal Sciences, University of Udine, Via Sondrio 2/A, Udine, 33100, Italy.
- Food Quality and Design Group, Wageningen University, Bornse Weilanden 9, Wageningen, 6708 WG, The Netherlands.
| |
Collapse
|
36
|
Mei Y, Li A, Zhao J, Li Y, Zhou Q, Yang M, Zhao M, Xu J, Li K, Yin G, Wu J, Xu Q. Disturbed glucose homeostasis and its increased allostatic load in response to individual, joint and fluctuating air pollutants exposure: Evidence from a longitudinal study in prediabetes. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 951:175498. [PMID: 39151627 DOI: 10.1016/j.scitotenv.2024.175498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/30/2024] [Accepted: 08/12/2024] [Indexed: 08/19/2024]
Abstract
We investigated the effect of individual, joint and fluctuating exposure to air pollution (PM2.5, BC, NO3-, NH4+, OM, SO42-, PM10, NO2, SO2, O3) on glucose metabolisms among prediabetes, and simultaneously explored the modifying effect of lifestyle. We conducted a longitudinal study among prediabetes during 2018-2022. Exposure windows within 60-days moving averages and their variabilities were calculated. FBG, insulin, HOMA-IR, HOMA-B, triglyceride glucose index (TyG), glucose insulin ratio (GI) and allostatic load of glucose homeostasis system (AL-GHS) was included. Linear mixed-effects model and BKMR were adopted to investigate the individual and overall effects, respectively. We also explored the preventive role of lifestyle. Individual air pollutant was associated with increased FBG, insulin, HOMA-IR, HOMA-B, TyG, and decreased GI. People with FBG ≥6.1 mmol/L were more susceptible. Air pollutants mixture were only associated with increased HOMA-B, and constituents have the highest group-PIP. Air pollutants variation also exert harmful effect. We observed similar diabetic effect on AL-GHS. Finally, the diabetic effect of air pollutants disappeared if participants adopt a favorable lifestyle. Our findings highlighted the importance of comprehensively assessing multiple air pollutants and their variations, focusing on metabolic health status in the early prevention of T2D, and adopting healthy lifestyle to mitigate such harmful effect.
Collapse
Affiliation(s)
- Yayuan Mei
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; Big Data Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100046, China; Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China
| | - Ang Li
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China
| | - Jiaxin Zhao
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China
| | - Yanbing Li
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China
| | - Quan Zhou
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China
| | - Ming Yang
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China
| | - Meiduo Zhao
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China
| | - Jing Xu
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China
| | - Kai Li
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China
| | - Guohuan Yin
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China
| | - Jingtao Wu
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China
| | - Qun Xu
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China.
| |
Collapse
|
37
|
Nie P, Hu L, Feng X, Xu H. Gut Microbiota Disorders and Metabolic Syndrome: Tales of a Crosstalk Process. Nutr Rev 2024:nuae157. [PMID: 39504479 DOI: 10.1093/nutrit/nuae157] [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] [Indexed: 11/08/2024] Open
Abstract
The microbiota in humans consists of trillions of microorganisms that are involved in the regulation of the gastrointestinal tract and immune and metabolic homeostasis. The gut microbiota (GM) has a prominent impact on the pathogenesis of metabolic syndrome (MetS). This process is reciprocal, constituting a crosstalk process between the GM and MetS. In this review, GM directly or indirectly inducing MetS via the host-microbial metabolic axis has been systematically reviewed. Additionally, the specifically altered GM in MetS are detailed in this review. Moreover, short-chain fatty acids (SCFAs), as unique gut microbial metabolites, have a remarkable effect on MetS, and the role of SCFAs in MetS-related diseases is highlighted to supplement the gaps in this area. Finally, the existing therapeutics are outlined, and the superiority and shortcomings of different therapeutic approaches are discussed, in hopes that this review can contribute to the development of potential treatment strategies.
Collapse
Affiliation(s)
- Penghui Nie
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Liehai Hu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Xiaoyan Feng
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Hengyi Xu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
- International Institute of Food Innovation Co., Ltd, Nanchang University, Nanchang 330200, China
| |
Collapse
|
38
|
Kryska A, Depciuch J, Krysa M, Paja W, Wosiak A, Nicoś M, Budzynska B, Sroka-Bartnicka A. Lipids balance as a spectroscopy marker of diabetes. Analysis of FTIR spectra by 2D correlation and machine learning analyses. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 320:124653. [PMID: 38901232 DOI: 10.1016/j.saa.2024.124653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 05/28/2024] [Accepted: 06/11/2024] [Indexed: 06/22/2024]
Abstract
The number of people suffering from type 2 diabetes has rapidly increased. Taking into account, that elevated intracellular lipid concentrations, as well as their metabolism, are correlated with diminished insulin sensitivity, in this study we would like to show lipids spectroscopy markers of diabetes. For this purpose, serum collected from rats (animal model of diabetes) was analyzed using Fourier Transformed Infrared-Attenuated Total Reflection (FTIR-ATR) spectroscopy. Analyzed spectra showed that rats with diabetes presented higher concentration of phospholipids and cholesterol in comparison with non-diabetic rats. Moreover, the analysis of second (IInd) derivative spectra showed no structural changes in lipids. Machine learning methods showed higher accuracy for IInd derivative spectra (from 65 % to 89 %) than for absorbance FTIR spectra (53-65 %). Moreover, it was possible to identify significant wavelength intervals from IInd derivative spectra using random forest-based feature selection algorithm, which further increased the accuracy of the classification (up to 92 % for phospholipid region). Moreover decision tree based on the selected features showed, that peaks at 1016 cm-1 and 2936 cm-1 can be good candidates of lipids marker of diabetes.
Collapse
Affiliation(s)
- Adrianna Kryska
- Independent Unit of Spectroscopy and Chemical Imaging, Faculty of Biomedical Sciences, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Joanna Depciuch
- Institute of Nuclear Physics, Polish Academy of Sciences, Walerego Eljasza - Radzikowskiego 152, 31-342 Kraków, Poland; Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodźki 1, Lublin 20-093, Poland
| | - Mikolaj Krysa
- Independent Unit of Spectroscopy and Chemical Imaging, Faculty of Biomedical Sciences, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Wiesław Paja
- Institute of Computer Science, University of Rzeszow, Pigonia 1, 35-310 Rzeszów, Poland
| | - Agnieszka Wosiak
- Institute of Information Technology, Lodz University of Technology, Politechniki 8, 93-590 Łódź, Poland
| | - Marcin Nicoś
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Jaczewskiego 8, 20-090 Lublin, Poland
| | - Barbara Budzynska
- Independent Laboratory of Behavioral Studies, Faculty of Biomedical Sciences, Medical University of Lublin, Chodzki 4a, 20-093 Lublin, Poland
| | - Anna Sroka-Bartnicka
- Independent Unit of Spectroscopy and Chemical Imaging, Faculty of Biomedical Sciences, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland.
| |
Collapse
|
39
|
Bechtel W, Bich L. Situating homeostasis in organisms: maintaining organization through time. J Physiol 2024; 602:6003-6020. [PMID: 39383321 DOI: 10.1113/jp286883] [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: 05/08/2024] [Accepted: 09/11/2024] [Indexed: 10/11/2024] Open
Abstract
Since it was inspired by Bernard and developed and named by Cannon, the concept of homeostasis has been invoked by many as the central theoretical framework for physiology. It has also been the target of numerous criticisms that have elicited the introduction of a plethora of alternative concepts. We argue that many of the criticisms actually target the more restrictive account of homeostasis advanced by the cyberneticists. What was crucial to Bernard and Cannon was a focus on the maintenance of the organism as the goal of physiological regulation. We analyse how Bernard's and Cannon's broad conception of what was required to maintain the organism was narrowed to negative feedback, characterized in terms of setpoints, by the cyberneticists and demonstrate how many of the alternative concepts challenge the role of setpoints - treating them as variable in light of circumstances or in anticipation of future circumstances, or as dispensable altogether. To support our analysis, we draw on the experimental and theoretical work on thermoregulation, a phenomenon that has been considered as a paradigmatic example of homeostasis and has been a common focus of those advancing alternative concepts. To integrate the insights advanced by the original proponents of homeostasis and the theorists proposing replacement notions we advance a framework in which regulation is viewed from the perspective of maintaining the organism.
Collapse
Affiliation(s)
- William Bechtel
- Department of Philosophy, University of California, San Diego, CA, USA
| | - Leonardo Bich
- Department of Philosophy, IAS-Research Centre for Life, Mind and Society, University of the Basque Country (UPV/EHU), Donostia-San Sebastian, Spain
| |
Collapse
|
40
|
Hamilton R, McCarthy OM, Bain SC, Bracken RM. Continuous measurement of interstitial glycaemia in professional female UCI world tour cyclists undertaking a 9-day cycle training camp. Eur J Sport Sci 2024; 24:1573-1582. [PMID: 39340462 PMCID: PMC11534661 DOI: 10.1002/ejsc.12201] [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: 11/15/2023] [Revised: 06/10/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024]
Abstract
Nine cyclists (age: 26 ± 5 years, height: 168 ± 5 cm and mass 58.5 ± 4.5 kg) were observed using continuous glucose monitoring devices throughout a training camp. Interstitial glucose [iG] data were captured via the Abbott libre sense biosensor (Abbott Laboratories) and paired with the Supersapiens software (TT1 Products Inc.). [iG] data were split into time ranges, that is, overall (24-hourly), day-time (06:00-23:59), night-time (00:00-05:59) and exercise. [iG] data were stratified into percentage of time, below range ([TBR] < 70 mg/dl), in range ([TIR] 70-140 mg/dl) and above range ([TAR] ≥ 141 mg/dl). Differences in diurnal and nocturnal data were analysed via repeated measures analysis of variance and paired t-tests where appropriate. p-value of ≤0.05 was accepted as significant. Riders spent an average of 3 ± 1% TAR, 93 ± 2% TIR and 8 ± 3% TBR. Mean 24 h [iG] was 93 ± 2 mg/dl with a coefficient of variation (CV) of 18 ± 1%. Mean (day: 95 ± 3 vs. night: 86 ± 3 mg/dl and p < 0.001) and CV (day: 18 ± 1 vs. night: 9 ± 1% and p < 0.001) in [iG] were higher during the day-time hours. TAR was greater during the day (day: 3 ± 1 vs. night: 0 ± 0% and p < 0.001) but TBR and TIR were similar. Glucose levels below the clinical range may have implications for those without diabetes and warrants further investigation.
Collapse
Affiliation(s)
- Ross Hamilton
- Applied Sport, Technology, Exercise and Medicine Research CentreSwansea UniversitySwanseaUK
| | - Olivia M. McCarthy
- Applied Sport, Technology, Exercise and Medicine Research CentreSwansea UniversitySwanseaUK
- Steno Diabetes Centre CopenhagenCopenhagen University HospitalHerlevDenmark
| | | | - Richard M. Bracken
- Applied Sport, Technology, Exercise and Medicine Research CentreSwansea UniversitySwanseaUK
- Faculty of Science and EngineeringHealth Technology and Solutions Interdisciplinary Research InstituteSwansea UniversitySwanseaUK
| |
Collapse
|
41
|
Tocharus C, Prum V, Sutheerawattananonda M. Oral Toxicity and Hypotensive Influence of Sericin-Derived Oligopeptides (SDOs) from Yellow Silk Cocoons of Bombyx mori in Rodent Studies. Foods 2024; 13:3505. [PMID: 39517289 PMCID: PMC11545120 DOI: 10.3390/foods13213505] [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: 10/11/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Sericin-derived oligopeptides (SDOs) from yellow silk cocoons exhibit antihypertensive and hypoglycemic properties in both in vitro and in vivo studies. This study investigated the acute toxicity of SDOs as a novel food for human consumption using female ICR mice and Wistar rats, as well as the chronic toxicity test on both sexes of Wistar rats. Clinical chemistry, hematology, and histopathological studies revealed that SDOs were safe for a single dose of 2000 mg kg-1 body weight (BW) and daily oral administration of 50, 100, and 200 mg kg-1 BW for six months. The chronic toxicity study additionally measured the rats' systolic blood pressure (SBP) and blood sugar monthly as they slowly aged. In the 2nd month for male rats and the 4th month for both sexes, SDOs had a significant hypotensive effect on Wistar rats' blood pressure, lowering it from 130 mmHg to a plateau at 110-115 mmHg. In contrast, the blood pressure of the control rats exceeded 140 mmHg after five months. Nonetheless, the hypoglycemic effect was not observed. Measurements of SBP and blood glucose in aged rats during chronic toxicity tests yielded insights beyond ordinary toxicity, including the health and fitness of the lab rats, perhaps resulting in novel discoveries or areas of study that justify the sacrifice of the animals' lives.
Collapse
Affiliation(s)
- Chainarong Tocharus
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Virakboth Prum
- School of Food Technology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand;
| | - Manote Sutheerawattananonda
- School of Food Technology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand;
| |
Collapse
|
42
|
Jeon J, Lee SW, Park HJ, Park YH, Kim TC, Lee S, Lee S, Van Kaer L, Hong S. Overexpression of Chromatin Remodeling Factor SRG3 Down-Regulates IL1β-Expressing M1 Macrophages and IL17-Producing T Cells in Adipose Tissues. Int J Mol Sci 2024; 25:11681. [PMID: 39519233 PMCID: PMC11546064 DOI: 10.3390/ijms252111681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
The SWItch3-related gene (SRG3) is a core component of ATP-dependent SWI/SNF complexes, which are crucial for regulating immune cell development and function (e.g., macrophages and CD4+ T cells), embryonic development, and non-immune cell differentiation. Notably, SRG3 overexpression has been shown to polarize macrophages in the central nervous system toward an anti-inflammatory M2 phenotype, thereby protecting against the development of experimental autoimmune encephalomyelitis in mice. However, the effect of SRG3 on immune responses in adipose tissues remains unclear. To address this issue, we examined the cellularity and inflammatory status of adipose tissue in B10.PL mice overexpressing the SRG3 gene under the ubiquitous β-actin promoter (SRG3β-actin). Interestingly, SRG3 overexpression significantly reduced adipocyte size in both white and brown adipose tissues, without affecting the overall adipose tissue weight. Such phenotypic effects might be associated with the improved glucose tolerance observed in SRG3β-actin B10.PL mice. Moreover, we found that SRG3 overexpression down-regulates IL1β-expressing M1 macrophages, leading to a significant decrease in the M1/M2 macrophage ratio. Additionally, SRG3β-actin B10.PL mice showed a dramatic reduction in neutrophils as well as IL1β- and IL17-producing T cells in adipose tissues. Taken together, our results indicate that SRG3 plays a vital role in maintaining immune homeostasis within adipose tissues.
Collapse
Affiliation(s)
- Jungmin Jeon
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul 05006, Republic of Korea; (J.J.); (H.J.P.); (Y.H.P.); (T.-C.K.); (S.L.); (S.L.)
| | - Sung Won Lee
- Department of Biomedical Laboratory Science, College of Health and Biomedical Services, Sangji University, Wonju 26339, Republic of Korea;
| | - Hyun Jung Park
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul 05006, Republic of Korea; (J.J.); (H.J.P.); (Y.H.P.); (T.-C.K.); (S.L.); (S.L.)
| | - Yun Hoo Park
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul 05006, Republic of Korea; (J.J.); (H.J.P.); (Y.H.P.); (T.-C.K.); (S.L.); (S.L.)
| | - Tae-Cheol Kim
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul 05006, Republic of Korea; (J.J.); (H.J.P.); (Y.H.P.); (T.-C.K.); (S.L.); (S.L.)
| | - Sujin Lee
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul 05006, Republic of Korea; (J.J.); (H.J.P.); (Y.H.P.); (T.-C.K.); (S.L.); (S.L.)
| | - Seyeong Lee
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul 05006, Republic of Korea; (J.J.); (H.J.P.); (Y.H.P.); (T.-C.K.); (S.L.); (S.L.)
| | - Luc Van Kaer
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA;
| | - Seokmann Hong
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul 05006, Republic of Korea; (J.J.); (H.J.P.); (Y.H.P.); (T.-C.K.); (S.L.); (S.L.)
| |
Collapse
|
43
|
Yang Y, Cheng H. Emerging Roles of ncRNAs in Type 2 Diabetes Mellitus: From Mechanisms to Drug Discovery. Biomolecules 2024; 14:1364. [PMID: 39595541 PMCID: PMC11592034 DOI: 10.3390/biom14111364] [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: 09/30/2024] [Revised: 10/23/2024] [Accepted: 10/26/2024] [Indexed: 11/28/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM), a high-incidence chronic metabolic disorder, has emerged as a global health issue, where most patients need lifelong medication. Gaining insights into molecular mechanisms involved in T2DM development is expected to provide novel strategies for clinical prevention and treatment. Growing evidence validates that non-coding RNAs (ncRNAs) including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs) function as crucial regulators in multiple biological processes of T2DM, inspiring various potential targets and drug candidates. In this review, we summarize the current understanding of ncRNA roles in T2DM and discuss the potential use of ncRNAs as targets and active molecules for drug discovery.
Collapse
Affiliation(s)
- Yue Yang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Hao Cheng
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
44
|
Izu GO, Mashele SS, Chukwuma CI. Investigating the Phytochemistry and Underlying Glycemic Control Mechanisms of Litchi chinensis Sonn. (Litchi) Peel Ethyl Acetate Extract in a Fructose/Streptozotocin Diabetic Model of Rats. Nutrients 2024; 16:3644. [PMID: 39519477 PMCID: PMC11547722 DOI: 10.3390/nu16213644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 10/19/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
The glycemic control potential and flavonoid profile of litchi have been documented for its hydroalcoholic extracts, while there is scarce information regarding its ethyl acetate extract. This study investigated the flavonoid profile, as well as the ameliorative potential and possible underlying mechanisms of litchi peel ethyl acetate extract on type 2 diabetes-related pathologies in a fructose/streptozotocin (STZ) model of diabetic rats. Sprague Dawley rats were induced with diabetes by administering 10% fructose for 2 weeks and a single i.p. injection of low-dose (40 mg/kg bw) STZ. Thereafter, the animals were orally administered with a low-dose (150 mg/kg bw) and high-dose (300 mg/kg bw) of the peel extract (LDPE and HDPE, respectively) and metformin (200 mg/kg bw). Compared to untreated diabetic rats (AUC = 1004 mg.h/dL), the HDPE significantly (p < 0.05) improved glucose tolerance (AUC = 847 mg.h/dL), which was statistically comparable (p ˃ 0.05) to the effect of metformin (AUC = 903 mg.h/dL). Serum insulin and pancreatic histology data showed that the STZ-induced pancreatic damage and insulin depletion was improved by the HDPE, which could be linked to the observed ameliorative effect of the extract on pancreatic lipid peroxidation and SOD and catalase activity. The extract further improved liver and muscle glycogen storage, as well as muscle hexokinase activity and Akt phosphorylation, suggesting that the extract exerts glycemic control by enhancing glycogen storage and modulating insulin-mediated signaling of glucose uptake and utilization. LC-MS data and documented reports suggest that flavonoids, such as epicatechin, cinnamtannin B2, procyanidin B5, and proanthocyanidin A2, are the possible influencing compounds. The ethyl acetate extract of litchi peel could be a source of bioactive flavonoids that can potentiate glycemic control in diabetes and mitigate oxidative stress-related pathologies.
Collapse
Affiliation(s)
- Gloria O. Izu
- Department of Health Sciences, Faculty of Health and Environmental Sciences, Central University of Technology, Bloemfontein 9301, Free State, South Africa;
- Centre for Quality of Health and Living (CQHL), Faculty of Health and Environmental Sciences, Central University of Technology, Private Bag X20539, Bloemfontein 9300, Free State, South Africa;
| | - Samson S. Mashele
- Centre for Quality of Health and Living (CQHL), Faculty of Health and Environmental Sciences, Central University of Technology, Private Bag X20539, Bloemfontein 9300, Free State, South Africa;
| | - Chika I. Chukwuma
- Centre for Quality of Health and Living (CQHL), Faculty of Health and Environmental Sciences, Central University of Technology, Private Bag X20539, Bloemfontein 9300, Free State, South Africa;
| |
Collapse
|
45
|
Ho KH, Barmaver SN, Hu R, Yagan M, Ahmed HK, Kaverina I, Gu G. Pancreatic islet α cells regulate microtubule stability in neighboring β cells to tune insulin secretion and induce functional heterogeneity in individual mouse and human islets. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.21.619544. [PMID: 39484371 PMCID: PMC11527287 DOI: 10.1101/2024.10.21.619544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
We have reported that the microtubule (MT) network in β cells attenuates this function by withdrawing insulin secretory granules (ISGs) away from the plasma membrane. Thus, high glucose-induced MT remodeling is required for robust glucose-stimulated insulin secretion (GSIS). We now show that α-cell secreted hormones, Gcg and/or Glp1, regulate the MT stability in β cells. Activating the receptors of Gcg or Glp1 (GcgR or Glp1R) with chemical agonists induces MT destabilization in β ells in the absence of high glucose. In contrast, inhibiting these receptors with antagonists attenuates high glucose-induced MT destabilization. Supporting the significance of this regulation, the MT networks in β cells of islets with higher α/β cell ratio are less stable than those with lower α/β cell ratio. Within each individual islet, β cells that are located close to α cells show faster MTs remodeling upon glucose stimulation than those away. Consequently, islets with higher α/β cell ratio secrete more insulin in response to high glucose and plasma membrane depolarization, which is recapitulated by direct Gcg stimulation. These combined results reveal a new MT-dependent pathway by which α cells, using Gcg and or Glp1-mediated paracrine signaling, tune β-cell secretion. In addition, the different α-β cell ratios in individual islets lead to their heterogeneous secretory responses, which may be important for handling secretory function needs under different physiological conditions. Highlights Gcg sensitizes glucose-induced MT remodeling in mouse and human β cellsMT density in single islets anti-correlates with α/β cell ratioGSIS levels in single islets positively correlate with α/β cell ratioDifferent α/β cell ratio contributes to heterogeneity of single islet GSIS.
Collapse
|
46
|
Tegegne BA, Adugna A, Yenet A, Yihunie Belay W, Yibeltal Y, Dagne A, Hibstu Teffera Z, Amare GA, Abebaw D, Tewabe H, Abebe RB, Zeleke TK. A critical review on diabetes mellitus type 1 and type 2 management approaches: from lifestyle modification to current and novel targets and therapeutic agents. Front Endocrinol (Lausanne) 2024; 15:1440456. [PMID: 39493778 PMCID: PMC11527681 DOI: 10.3389/fendo.2024.1440456] [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: 05/29/2024] [Accepted: 10/02/2024] [Indexed: 11/05/2024] Open
Abstract
Diabetes mellitus (DM) has emerged as an international health epidemic due to its rapid rise in prevalence. Consequently, scientists and or researchers will continue to find novel, safe, effective, and affordable anti-diabetic medications. The goal of this review is to provide a thorough overview of the role that lifestyle changes play in managing diabetes, as well as the standard medications that are currently being used to treat the condition and the most recent advancements in the development of novel medical treatments that may be used as future interventions for the disease. A literature search was conducted using research databases such as PubMed, Web of Science, Scopus, ScienceDirect, Wiley Online Library, Google Scholar, etc. Data were then abstracted from these publications using words or Phrases like "pathophysiology of diabetes", "Signe and symptoms of diabetes", "types of diabetes", "major risk factors and complication of diabetes", "diagnosis of diabetes", "lifestyle modification for diabetes", "current antidiabetic agents", and "novel drugs and targets for diabetes management" that were published in English and had a strong scientific foundation. Special emphasis was given to the importance of lifestyle modification, as well as current, novel, and emerging/promising drugs and targets helpful for the management of both T1DM and T2DM.
Collapse
Affiliation(s)
- Bantayehu Addis Tegegne
- Department of Pharmacy, College of Medicine and Health Science, Debre Markos University, Debre Markos, Ethiopia
| | - Adane Adugna
- Department of Medical Laboratory Science, College of Medicine and Health Science, Debre Markos University, Debre Markos, Ethiopia
| | - Aderaw Yenet
- Department of Pharmacy, College of Medicine and Health Science, Debre Markos University, Debre Markos, Ethiopia
| | - Wubetu Yihunie Belay
- Department of Pharmacy, College of Medicine and Health Science, Debre Markos University, Debre Markos, Ethiopia
| | - Yared Yibeltal
- Department of Pharmacy, College of Medicine and Health Science, Debre Markos University, Debre Markos, Ethiopia
| | - Abebe Dagne
- Department of Pharmacy, College of Medicine and Health Science, Debre Markos University, Debre Markos, Ethiopia
| | - Zigale Hibstu Teffera
- Department of Medical Laboratory Science, College of Medicine and Health Science, Debre Markos University, Debre Markos, Ethiopia
| | - Gashaw Azanaw Amare
- Department of Medical Laboratory Science, College of Medicine and Health Science, Debre Markos University, Debre Markos, Ethiopia
| | - Desalegn Abebaw
- Department of Medical Laboratory Science, College of Medicine and Health Science, Debre Markos University, Debre Markos, Ethiopia
| | - Haymanot Tewabe
- Department of Medical Laboratory Science, College of Medicine and Health Science, Debre Markos University, Debre Markos, Ethiopia
| | - Rahel Belete Abebe
- Department of Clinical Pharmacy, School of Pharmacy, College of Medicine and Health Science, University of Gondar, Gondar, Ethiopia
| | - Tirsit Ketsela Zeleke
- Department of Pharmacy, College of Medicine and Health Science, Debre Markos University, Debre Markos, Ethiopia
| |
Collapse
|
47
|
Nxumalo MB, Ntanzi N, Kumalo HM, Khan RB. Mitigating Hyperglycaemic Oxidative Stress in HepG2 Cells: The Role of Carica papaya Leaf and Root Extracts in Promoting Glucose Uptake and Antioxidant Defence. Nutrients 2024; 16:3496. [PMID: 39458491 PMCID: PMC11510471 DOI: 10.3390/nu16203496] [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: 09/05/2024] [Revised: 09/30/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Diabetes often goes undiagnosed, with 60% of people in Africa unaware of their condition. Type 2 diabetes mellitus (T2DM) is associated with insulin resistance and is treated with metformin, despite the undesirable side effects. Medicinal plants with therapeutic potential, such as Carica papaya, have shown promising anti-diabetic properties. This study explored the role of C. papaya leaf and root extracts compared to metformin in reducing hyperglycaemia-induced oxidative stress and their impact on liver function using HepG2 as a reference. Methods: The cytotoxicity was assessed through the MTT assay. At the same time, glucose uptake and metabolism (ATP and ∆Ψm) in HepG2 cells treated with C. papaya aqueous leaf and root extract were evaluated using a luminometry assay. Additionally, antioxidant properties (SOD2, GPx1, GSH, and Nrf2) were measured using qPCR and Western blot following the detection of MDA, NO, and iNOS, indicators of free radicals. Results: The MTT assay showed that C. papaya extracts did not exhibit toxicity in HepG2 cells and enhanced glucose uptake compared to the hyperglycaemic control (HGC) and metformin. The glucose levels in C. papaya-treated cells increased ATP production (p < 0.05), while the ∆Ψm was significantly increased in HGR1000-treated cells (p < 0.05). Furthermore, C. papaya leaf extract upregulated GPx1 (p < 0.05), GSH, and Nrf2 gene (p < 0.05), while SOD2 and Nrf2 proteins were reduced (p > 0.05), ultimately lowering ROS (p > 0.05). Contrarily, the root extract stimulated SOD2 (p > 0.05), GPx1 (p < 0.05), and GSH levels (p < 0.05), reducing Nrf2 gene and protein expression (p < 0.05) and resulting in high MDA levels (p < 0.05). Additionally, the extracts elevated NO levels and iNOS expression (p < 0.05), suggesting potential RNS activation. Conclusion: Taken together, the leaf extract stimulated glucose metabolism and triggered ROS production, producing a strong antioxidant response that was more effective than the root extract and metformin. However, the root extract, particularly at high concentrations, was less effective at neutralising free radicals as it did not stimulate Nrf2 production, but it did maintain elevated levels of SOD2, GSH, and GPx1 antioxidants.
Collapse
Affiliation(s)
- Mthokozisi Bongani Nxumalo
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa; (N.N.); (H.M.K.); (R.B.K.)
| | | | | | | |
Collapse
|
48
|
Yang L, Qiao S, Zhang G, Lu A, Li F. Inflammatory Processes: Key Mediators of Oncogenesis and Progression in Pancreatic Ductal Adenocarcinoma (PDAC). Int J Mol Sci 2024; 25:10991. [PMID: 39456771 PMCID: PMC11506938 DOI: 10.3390/ijms252010991] [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: 09/25/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Associations between inflammation and cancer were first discovered approximately 160 years ago by Rudolf Virchow, who observed that tumors were infiltrated with inflammatory cells, and defined inflammation as a pathological condition. Inflammation has now emerged as one of the key mediators in oncogenesis and tumor progression, including pancreatic ductal adenocarcinoma (PDAC). However, the role of inflammatory processes in cancers is complicated and controversial, and the detailed regulatory mechanisms are still unclear. This review elucidates the dynamic interplay between inflammation and immune regulation, microenvironment alteration, metabolic reprogramming, and microbiome risk factors in PDAC, committing to exploring a deeper understanding of the role of crucial inflammatory pathways and molecules for providing insights into therapeutic strategies.
Collapse
Affiliation(s)
- Liu Yang
- Shum Yiu Foon Shum Bik Chuen Memorial Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; (L.Y.); (S.Q.)
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China;
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Shuangying Qiao
- Shum Yiu Foon Shum Bik Chuen Memorial Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; (L.Y.); (S.Q.)
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China;
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Ge Zhang
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China;
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Aiping Lu
- Shum Yiu Foon Shum Bik Chuen Memorial Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; (L.Y.); (S.Q.)
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China;
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Fangfei Li
- Shum Yiu Foon Shum Bik Chuen Memorial Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; (L.Y.); (S.Q.)
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China;
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| |
Collapse
|
49
|
Kim TY, De R, Choi I, Kim H, Hahn SK. Multifunctional nanomaterials for smart wearable diabetic healthcare devices. Biomaterials 2024; 310:122630. [PMID: 38815456 DOI: 10.1016/j.biomaterials.2024.122630] [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: 01/11/2024] [Accepted: 05/19/2024] [Indexed: 06/01/2024]
Abstract
Wearable diabetic healthcare devices have attracted great attention for real-time continuous glucose monitoring (CGM) using biofluids such as tears, sweat, saliva, and interstitial fluid via noninvasive ways. In response to the escalating global demand for CGM, these devices enable proactive management and intervention of diabetic patients with incorporated drug delivery systems (DDSs). In this context, multifunctional nanomaterials can trigger the development of innovative sensing and management platforms to facilitate real-time selective glucose monitoring with remarkable sensitivity, on-demand drug delivery, and wireless power and data transmission. The seamless integration into wearable devices ensures patient's compliance. This comprehensive review evaluates the multifaceted roles of these materials in wearable diabetic healthcare devices, comparing their glucose sensing capabilities with conventionally available glucometers and CGM devices, and finally outlines the merits, limitations, and prospects of these devices. This review would serve as a valuable resource, elucidating the intricate functions of nanomaterials for the successful development of advanced wearable devices in diabetes management.
Collapse
Affiliation(s)
- Tae Yeon Kim
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk, 37673, South Korea
| | - Ranjit De
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk, 37673, South Korea
| | - Inhoo Choi
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk, 37673, South Korea
| | - Hyemin Kim
- Department of Cosmetics Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, South Korea.
| | - Sei Kwang Hahn
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk, 37673, South Korea.
| |
Collapse
|
50
|
Abo SMC, Layton AT. Modeling sex-specific whole-body metabolic responses to feeding and fasting. Comput Biol Med 2024; 181:109024. [PMID: 39178806 DOI: 10.1016/j.compbiomed.2024.109024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/27/2024] [Accepted: 08/11/2024] [Indexed: 08/26/2024]
Abstract
Men generally favor carbohydrate metabolism, while women lean towards lipid metabolism, resulting in significant sex-based differences in energy oxidation across various metabolic states such as fasting and feeding. These differences are influenced by body composition and inherent metabolic fluxes, including increased lipolysis rates in women. However, understanding how sex influences organ-specific metabolism and systemic manifestations remains incomplete. To address these gaps, we developed a sex-specific, whole-body metabolic model for feeding and fasting scenarios in healthy young adults. Our model integrates organ metabolism with whole-body responses to mixed meals, particularly high-carbohydrate and high-fat meals. Our predictions suggest that differences in liver and adipose tissue nutrient storage and oxidation patterns drive systemic metabolic disparities. We propose that sex differences in fasting hepatic glucose output may result from the different handling of free fatty acids, glycerol, and glycogen. We identified a metabolic pathway, possibly more prevalent in female livers, redirecting lipids towards carbohydrate metabolism to support hepatic glucose production. This mechanism is facilitated by the TG-FFA cycle between adipose tissue and the liver. Incorporating sex-specific data into multi-scale frameworks offers insights into how sex modulates human metabolism.
Collapse
Affiliation(s)
- Stéphanie M C Abo
- Department of Applied Mathematics, University of Waterloo, 200 University Ave W, Waterloo, N2L 3G1, Ontario, Canada.
| | - Anita T Layton
- Department of Applied Mathematics, University of Waterloo, 200 University Ave W, Waterloo, N2L 3G1, Ontario, Canada; Cheriton School of Computer Science, Department of Biology, and School of Pharmacy, 200 University Ave W, Waterloo, N2L 3G1, Ontario, Canada.
| |
Collapse
|