Pub Date : 2025-01-08eCollection Date: 2024-01-01DOI: 10.3389/fendo.2024.1475958
Qianqian Liang, Yide Wang, Zheng Li
Background: Diabetes and chronic obstructive pulmonary disease (COPD) are prominent global health challenges, each imposing significant burdens on affected individuals, healthcare systems, and society. However, the specific molecular mechanisms supporting their interrelationship have not been fully defined.
Methods: We identified the differentially expressed genes (DEGs) of COPD and diabetes from multi-center patient cohorts, respectively. Through cross-analysis, we identified the shared DEGs of COPD and diabetes, and investigated alterations of signaling pathways using Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and gene set enrichment analysis (GSEA). By using weighted gene correlation network analysis (WGCNA), key gene modules for COPD and diabetes were identified, and various machine learning algorithms were employed to identify shared biomarkers. Using xCell, we investigated the relationship between shared biomarkers and immune infiltration in diabetes and COPD. Single-cell sequencing, clinical samples, and animal models were used to confirm the robustness of shared biomarkers.
Results: Cross-analysis identified 186 shared DEGs between diabetes and COPD patients. Functional enrichment results demonstrate that metabolic and immune-related pathways are common features altered in both diabetes and COPD patients. WGCNA identified 526 genes from key gene modules in COPD and diabetes. Multiple machine learning algorithms identified 4 shared biomarkers for COPD and diabetes, including CADPS, EDNRB, THBS4 and TMEM27. Finally, the 4 shared biomarkers were validated in single-cell sequencing data, clinical samples, and animal models, and their expression changes were consistent with the results of bioinformatic analysis.
Conclusions: Through comprehensive bioinformatics analysis, we revealed the potential connection between diabetes and COPD, providing a theoretical basis for exploring the common regulatory genes.
{"title":"Comprehensive bioinformatics analysis identifies metabolic and immune-related diagnostic biomarkers shared between diabetes and COPD using multi-omics and machine learning.","authors":"Qianqian Liang, Yide Wang, Zheng Li","doi":"10.3389/fendo.2024.1475958","DOIUrl":"10.3389/fendo.2024.1475958","url":null,"abstract":"<p><strong>Background: </strong>Diabetes and chronic obstructive pulmonary disease (COPD) are prominent global health challenges, each imposing significant burdens on affected individuals, healthcare systems, and society. However, the specific molecular mechanisms supporting their interrelationship have not been fully defined.</p><p><strong>Methods: </strong>We identified the differentially expressed genes (DEGs) of COPD and diabetes from multi-center patient cohorts, respectively. Through cross-analysis, we identified the shared DEGs of COPD and diabetes, and investigated alterations of signaling pathways using Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and gene set enrichment analysis (GSEA). By using weighted gene correlation network analysis (WGCNA), key gene modules for COPD and diabetes were identified, and various machine learning algorithms were employed to identify shared biomarkers. Using xCell, we investigated the relationship between shared biomarkers and immune infiltration in diabetes and COPD. Single-cell sequencing, clinical samples, and animal models were used to confirm the robustness of shared biomarkers.</p><p><strong>Results: </strong>Cross-analysis identified 186 shared DEGs between diabetes and COPD patients. Functional enrichment results demonstrate that metabolic and immune-related pathways are common features altered in both diabetes and COPD patients. WGCNA identified 526 genes from key gene modules in COPD and diabetes. Multiple machine learning algorithms identified 4 shared biomarkers for COPD and diabetes, including CADPS, EDNRB, THBS4 and TMEM27. Finally, the 4 shared biomarkers were validated in single-cell sequencing data, clinical samples, and animal models, and their expression changes were consistent with the results of bioinformatic analysis.</p><p><strong>Conclusions: </strong>Through comprehensive bioinformatics analysis, we revealed the potential connection between diabetes and COPD, providing a theoretical basis for exploring the common regulatory genes.</p>","PeriodicalId":12447,"journal":{"name":"Frontiers in Endocrinology","volume":"15 ","pages":"1475958"},"PeriodicalIF":3.9,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11750655/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143023001","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-08eCollection Date: 2024-01-01DOI: 10.3389/fendo.2024.1507657
Hui Shi
Purpose: This study aimed to investigate the alterations in diabetes risk associated with sarcopenia and insufficient physical activity, as well as the demographic shifts within the diabetic population.
Method: Utilizing pertinent data from the National Health and Nutrition Examination Survey (NHANES) database spanning 2011 to 2018, the criteria for sarcopenia were established by the Foundation for the National Institutes of Health. These criteria were calculated using lean body mass data in conjunction with body mass index data. Physical activity levels were assessed using the PAQ questionnaire from the NHANES database. The presence of diabetes was determined through the DIQ questionnaire and the laboratory examination within the NHANES database. The analysis was performed using multivariable logistic regression.
Result: The prevalence of both sarcopenia and insufficient physical activity in the diabetic population was 188% greater than in the non-diabetic population. Sarcopenia and insufficient physical activity were positively correlated with an increased risk of diabetes onset, demonstrating a 1.45-fold heightened risk when both conditions were present (OR=2.45,95%CI,1.35-4.44,P<0.05). This combined effect was significantly greater than the risk associated with sarcopenia alone (OR=1.84,95%CI,1.09-3.11,P<0.05) or insufficient physical activity alone (OR=1.55,95%CI,1.11-2.15,P<0.05).
Conclusion: A synergistic relationship exists between sarcopenia and insufficient physical activity, resulting in a markedly elevated risk of diabetes when both conditions are present concurrently. Therefore, comprehensive diabetes management strategies should prioritize populations exhibiting both sarcopenia and insufficient physical activity.
{"title":"Unraveling the dual threat: sarcopenia and insufficient physical activity in diabetes risk.","authors":"Hui Shi","doi":"10.3389/fendo.2024.1507657","DOIUrl":"10.3389/fendo.2024.1507657","url":null,"abstract":"<p><strong>Purpose: </strong>This study aimed to investigate the alterations in diabetes risk associated with sarcopenia and insufficient physical activity, as well as the demographic shifts within the diabetic population.</p><p><strong>Method: </strong>Utilizing pertinent data from the National Health and Nutrition Examination Survey (NHANES) database spanning 2011 to 2018, the criteria for sarcopenia were established by the Foundation for the National Institutes of Health. These criteria were calculated using lean body mass data in conjunction with body mass index data. Physical activity levels were assessed using the PAQ questionnaire from the NHANES database. The presence of diabetes was determined through the DIQ questionnaire and the laboratory examination within the NHANES database. The analysis was performed using multivariable logistic regression.</p><p><strong>Result: </strong>The prevalence of both sarcopenia and insufficient physical activity in the diabetic population was 188% greater than in the non-diabetic population. Sarcopenia and insufficient physical activity were positively correlated with an increased risk of diabetes onset, demonstrating a 1.45-fold heightened risk when both conditions were present (OR=2.45,95%CI,1.35-4.44,P<0.05). This combined effect was significantly greater than the risk associated with sarcopenia alone (OR=1.84,95%CI,1.09-3.11,P<0.05) or insufficient physical activity alone (OR=1.55,95%CI,1.11-2.15,P<0.05).</p><p><strong>Conclusion: </strong>A synergistic relationship exists between sarcopenia and insufficient physical activity, resulting in a markedly elevated risk of diabetes when both conditions are present concurrently. Therefore, comprehensive diabetes management strategies should prioritize populations exhibiting both sarcopenia and insufficient physical activity.</p>","PeriodicalId":12447,"journal":{"name":"Frontiers in Endocrinology","volume":"15 ","pages":"1507657"},"PeriodicalIF":3.9,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11750674/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143022498","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-08eCollection Date: 2024-01-01DOI: 10.3389/fendo.2024.1509501
Sheng Zhang, Zhongzhou Su, Xianqiang Wen
<p><strong>Background: </strong>Abnormal thyroid hormone levels may occur in critical illness, which may have an interactive relationship with inflammatory reaction. At present, the relationship between triiodothyronine (T3)/thyroxine (T4) ratio and inflammatory indicators and all-cause mortality of stroke survivors is still unclear.</p><p><strong>Methods: </strong>We obtained the relevant data of the respondents from 2007 to 2012 through the National Health and Nutrition Examination Survey (NHANES) database for statistical analysis. The ratio of T3/T4, a continuous variable, is transformed into three groups of classified variables, namely Q1, Q2 and Q3. The relationship between T3/T4 ratio and mortality was analyzed by Log-Rank test and K-M survival curve. Pearson correlation analysis was used to analyze the correlation between T3/T4 ratio and white blood cell (WBC), Neutrophil-to-Lymphocyte Ratio (NLR), systemic immune-inflammation index (SII) and neutrophil percentage-to-albumin ratio (NPAR). Cox univariate and multivariate analysis was used to identify independent risk factors for all-cause mortality in stroke survivors and a nomogram was drawn. Restricted cubic spline (RCS) curve was drawn to determine whether there was a linear relationship between T3/T4 ratio and mortality and the best cut-off value. Subgroup analysis showed the difference between the T3/T4 ratio and all-cause mortality among subgroups and a forest plot was drawn. The mediation effect analysis was used to analyze whether the ratio of T3/T4 could mediate the survival time through inflammatory indicators.</p><p><strong>Results: </strong>According to the inclusion and exclusion criteria, a total of 267 people were included in the study, with a mortality rate of 49.06% (131/267), an average survival time of 111.22 ± 3.19 months, and a median survival time of 130 ± 11.27 months. The Log-Rank test and K-M survival curve showed that there were statistical differences among the Q1, Q2, and Q3 groups of the T3/T4 ratio (<i>x</i> <sup>2</sup> = 16.32, <i>p</i><0.001), and the lower the T3/T4 level, the shorter the survival time. Pearson correlation analysis showed that the T3/T4 ratio had a linear relationship with NLR, SII, and NPAR, and only had a weak correlation with NPAR (<i>r</i> = -0.31, <i>p</i><0.001). Cox univariate analysis showed that age, marital status, race, cancer, T3/T4 ratio, NPAR and all-cause mortality were related. Multivariate regression analysis showed that age ≥ 60 years, race of non-Hispanic black, low T3/T4 ratio (<i>p</i> = 0.014, <i>HR</i> = 0.92, 95% <i>CI</i> = 0.87~0.98) and high NPAR (<i>p</i> = 0.009, <i>HR</i> = 2.50, 95% <i>CI</i> = 1.26~4.99) were independent risk factors for all-cause mortality. The RCS curve shows that the ratio of T3/T4 is linearly correlated with mortality, and the optimal cutoff value of T3/T4 is 12.97. Subgroup analysis showed that T3/T4 ratio is more likely to affect the survival of stroke survivors with BMI 18.5~28. Mediat
{"title":"Association of T3/T4 ratio with inflammatory indicators and all-cause mortality in stroke survivors.","authors":"Sheng Zhang, Zhongzhou Su, Xianqiang Wen","doi":"10.3389/fendo.2024.1509501","DOIUrl":"10.3389/fendo.2024.1509501","url":null,"abstract":"<p><strong>Background: </strong>Abnormal thyroid hormone levels may occur in critical illness, which may have an interactive relationship with inflammatory reaction. At present, the relationship between triiodothyronine (T3)/thyroxine (T4) ratio and inflammatory indicators and all-cause mortality of stroke survivors is still unclear.</p><p><strong>Methods: </strong>We obtained the relevant data of the respondents from 2007 to 2012 through the National Health and Nutrition Examination Survey (NHANES) database for statistical analysis. The ratio of T3/T4, a continuous variable, is transformed into three groups of classified variables, namely Q1, Q2 and Q3. The relationship between T3/T4 ratio and mortality was analyzed by Log-Rank test and K-M survival curve. Pearson correlation analysis was used to analyze the correlation between T3/T4 ratio and white blood cell (WBC), Neutrophil-to-Lymphocyte Ratio (NLR), systemic immune-inflammation index (SII) and neutrophil percentage-to-albumin ratio (NPAR). Cox univariate and multivariate analysis was used to identify independent risk factors for all-cause mortality in stroke survivors and a nomogram was drawn. Restricted cubic spline (RCS) curve was drawn to determine whether there was a linear relationship between T3/T4 ratio and mortality and the best cut-off value. Subgroup analysis showed the difference between the T3/T4 ratio and all-cause mortality among subgroups and a forest plot was drawn. The mediation effect analysis was used to analyze whether the ratio of T3/T4 could mediate the survival time through inflammatory indicators.</p><p><strong>Results: </strong>According to the inclusion and exclusion criteria, a total of 267 people were included in the study, with a mortality rate of 49.06% (131/267), an average survival time of 111.22 ± 3.19 months, and a median survival time of 130 ± 11.27 months. The Log-Rank test and K-M survival curve showed that there were statistical differences among the Q1, Q2, and Q3 groups of the T3/T4 ratio (<i>x</i> <sup>2</sup> = 16.32, <i>p</i><0.001), and the lower the T3/T4 level, the shorter the survival time. Pearson correlation analysis showed that the T3/T4 ratio had a linear relationship with NLR, SII, and NPAR, and only had a weak correlation with NPAR (<i>r</i> = -0.31, <i>p</i><0.001). Cox univariate analysis showed that age, marital status, race, cancer, T3/T4 ratio, NPAR and all-cause mortality were related. Multivariate regression analysis showed that age ≥ 60 years, race of non-Hispanic black, low T3/T4 ratio (<i>p</i> = 0.014, <i>HR</i> = 0.92, 95% <i>CI</i> = 0.87~0.98) and high NPAR (<i>p</i> = 0.009, <i>HR</i> = 2.50, 95% <i>CI</i> = 1.26~4.99) were independent risk factors for all-cause mortality. The RCS curve shows that the ratio of T3/T4 is linearly correlated with mortality, and the optimal cutoff value of T3/T4 is 12.97. Subgroup analysis showed that T3/T4 ratio is more likely to affect the survival of stroke survivors with BMI 18.5~28. Mediat","PeriodicalId":12447,"journal":{"name":"Frontiers in Endocrinology","volume":"15 ","pages":"1509501"},"PeriodicalIF":3.9,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11750665/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143022995","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-08eCollection Date: 2024-01-01DOI: 10.3389/fendo.2024.1479764
Kathryn Flynn, Jennifer Hatfield, Kevin Brown, Nicole Vietor, Thanh Hoang
Diabetes insipidus (DI) is a rare endocrine disease involving antidiuretic hormone (ADH), encompassing both central and nephrogenic causes. Inability to respond to or produce ADH leads to inability of the kidneys to reabsorb water, resulting in hypotonic polyuria and, if lack of hydration, hypernatremia. DI cannot be cured and is an unfamiliar disease process to many clinicians. This diagnosis must be distinguished from primary polydipsia and other causes of hypotonic polyuria. The main branchpoints in pathophysiology depend on the level of ADH pathology: the brain or the kidneys. Prompt diagnosis and treatment are critical as DI can cause substantial morbidity and mortality. The gold standard for diagnosis is a water deprivation test followed by desmopressin administration. There is promising research regarding a new surrogate marker of ADH called copeptin, which may simplify and improve the accuracy in diagnosing DI in the future. Patients with DI require adequate access to water, and there are nuances on treatment approaches depending on whether a patient is diagnosed with central or nephrogenic DI. This article describes a stepwise approach to recognition, diagnosis, and treatment of DI.
{"title":"Central and nephrogenic diabetes insipidus: updates on diagnosis and management.","authors":"Kathryn Flynn, Jennifer Hatfield, Kevin Brown, Nicole Vietor, Thanh Hoang","doi":"10.3389/fendo.2024.1479764","DOIUrl":"10.3389/fendo.2024.1479764","url":null,"abstract":"<p><p>Diabetes insipidus (DI) is a rare endocrine disease involving antidiuretic hormone (ADH), encompassing both central and nephrogenic causes. Inability to respond to or produce ADH leads to inability of the kidneys to reabsorb water, resulting in hypotonic polyuria and, if lack of hydration, hypernatremia. DI cannot be cured and is an unfamiliar disease process to many clinicians. This diagnosis must be distinguished from primary polydipsia and other causes of hypotonic polyuria. The main branchpoints in pathophysiology depend on the level of ADH pathology: the brain or the kidneys. Prompt diagnosis and treatment are critical as DI can cause substantial morbidity and mortality. The gold standard for diagnosis is a water deprivation test followed by desmopressin administration. There is promising research regarding a new surrogate marker of ADH called copeptin, which may simplify and improve the accuracy in diagnosing DI in the future. Patients with DI require adequate access to water, and there are nuances on treatment approaches depending on whether a patient is diagnosed with central or nephrogenic DI. This article describes a stepwise approach to recognition, diagnosis, and treatment of DI.</p>","PeriodicalId":12447,"journal":{"name":"Frontiers in Endocrinology","volume":"15 ","pages":"1479764"},"PeriodicalIF":3.9,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11750692/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143022999","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-08eCollection Date: 2024-01-01DOI: 10.3389/fendo.2024.1542771
Suset Dueñas-Disotuar, Ana Piñar-Gutiérrez, Irene de Lara-Rodríguez, Julia Sastre-Marcos, Emma Anda-Apiñániz, Amelia Oleaga-Alday, J C Galofré, Aida Orois, Victoria Alcázar-Lázaro, Laia Martínez-Guasch, Cecilia Sánchez-Ragnarsson, María Ángeles Gálvez-Moreno, Cristina Familiar-Casado, Tomás Martín-Hernández, Ana R Romero-Lluch
[This corrects the article DOI: 10.3389/fendo.2024.1466245.].
{"title":"Corrigendum: Bone metastasis in differentiated thyroid cancer: Spanish multicenter study of clinical characteristics, survival and prognostic factors.","authors":"Suset Dueñas-Disotuar, Ana Piñar-Gutiérrez, Irene de Lara-Rodríguez, Julia Sastre-Marcos, Emma Anda-Apiñániz, Amelia Oleaga-Alday, J C Galofré, Aida Orois, Victoria Alcázar-Lázaro, Laia Martínez-Guasch, Cecilia Sánchez-Ragnarsson, María Ángeles Gálvez-Moreno, Cristina Familiar-Casado, Tomás Martín-Hernández, Ana R Romero-Lluch","doi":"10.3389/fendo.2024.1542771","DOIUrl":"https://doi.org/10.3389/fendo.2024.1542771","url":null,"abstract":"<p><p>[This corrects the article DOI: 10.3389/fendo.2024.1466245.].</p>","PeriodicalId":12447,"journal":{"name":"Frontiers in Endocrinology","volume":"15 ","pages":"1542771"},"PeriodicalIF":3.9,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11751468/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143022139","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-08eCollection Date: 2024-01-01DOI: 10.3389/fendo.2024.1509445
Shimiao Bu, Jiang-Yue Ling, Xiaojun Wu, Liting Zhang, Xiangyu Shi, Lang Huang, Zheng Zhao, Ying Yang, Zongqin Xiang, Yong U Liu, Yufeng Liu, Yuehong Zhang
Objective: To explore the differential gene expression in peripheral blood immune cells of individuals with type 2 diabetes mellitus (DM), comparing those with and without non-proliferative diabetic retinopathy (NPDR).
Methods: From a pool of 126 potential participants, 60 were selected for detailed analysis. This group included 12 healthy donors (HDs), 22 individuals with DM, and 26 with NPDR. We analyzed peripheral blood mononuclear cells (PBMCs) using RNA sequencing and quantitative PCR (qPCR) to pinpoint differentially expressed genes (DEGs). Western blot and flow cytometry were also employed to evaluate the protein expression of specific genes.
Results: In patients with NPDR compared to those with DM alone, MerTK-a gene implicated in inherited retinal dystrophies due to its mutations-was notably downregulated in PBMCs. Through flow cytometry, we assessed the protein levels and cellular distribution of MerTK, finding a predominant expression in monocytes and myeloid-derived suppressor cells (MDSCs), with a marked reduction in CD4+ and CD8+ T cells, as well as in natural killer T (NKT) cells. Patients with DM demonstrated a significant deviation in the PBMCs composition, particularly in B cells, CD4+ T cells, and NK cells, when compared to HDs.
Conclusions: The study indicates that MerTK expression in T cells within PBMCs could act as a viable blood biomarker for NPDR risk in patients with DM. Furthermore, the regulation of T cells by MerTK might represent a critical pathway through which DM evolves into NPDR.
{"title":"Downregulation of MerTK in circulating T cells of patients with non-proliferative diabetic retinopathy.","authors":"Shimiao Bu, Jiang-Yue Ling, Xiaojun Wu, Liting Zhang, Xiangyu Shi, Lang Huang, Zheng Zhao, Ying Yang, Zongqin Xiang, Yong U Liu, Yufeng Liu, Yuehong Zhang","doi":"10.3389/fendo.2024.1509445","DOIUrl":"10.3389/fendo.2024.1509445","url":null,"abstract":"<p><strong>Objective: </strong>To explore the differential gene expression in peripheral blood immune cells of individuals with type 2 diabetes mellitus (DM), comparing those with and without non-proliferative diabetic retinopathy (NPDR).</p><p><strong>Methods: </strong>From a pool of 126 potential participants, 60 were selected for detailed analysis. This group included 12 healthy donors (HDs), 22 individuals with DM, and 26 with NPDR. We analyzed peripheral blood mononuclear cells (PBMCs) using RNA sequencing and quantitative PCR (qPCR) to pinpoint differentially expressed genes (DEGs). Western blot and flow cytometry were also employed to evaluate the protein expression of specific genes.</p><p><strong>Results: </strong>In patients with NPDR compared to those with DM alone, MerTK-a gene implicated in inherited retinal dystrophies due to its mutations-was notably downregulated in PBMCs. Through flow cytometry, we assessed the protein levels and cellular distribution of MerTK, finding a predominant expression in monocytes and myeloid-derived suppressor cells (MDSCs), with a marked reduction in CD4+ and CD8+ T cells, as well as in natural killer T (NKT) cells. Patients with DM demonstrated a significant deviation in the PBMCs composition, particularly in B cells, CD4+ T cells, and NK cells, when compared to HDs.</p><p><strong>Conclusions: </strong>The study indicates that MerTK expression in T cells within PBMCs could act as a viable blood biomarker for NPDR risk in patients with DM. Furthermore, the regulation of T cells by MerTK might represent a critical pathway through which DM evolves into NPDR.</p>","PeriodicalId":12447,"journal":{"name":"Frontiers in Endocrinology","volume":"15 ","pages":"1509445"},"PeriodicalIF":3.9,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11750652/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143022084","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-08eCollection Date: 2024-01-01DOI: 10.3389/fendo.2024.1414508
Chunmei Li, Qiongzhu Huang, Yanqian Zhuang, Pin Chen, Yiyang Lin
Objective: Type 2 diabetes mellitus (T2DM) is a major cause of atherosclerosis, as well as an independent risk factor of cardiovascular adverse events. We aimed to evaluate the association of serum Meteorin-like protein (Metrnl) level with carotid atherosclerosis as determined by carotid intima-media thickness (CIMT) status in subjects with T2DM.
Methods: This cross-sectional study included 83 T2DM subjects without pre-existing cardiovascular diseases. CIMT was measured by color doppler ultrasonography, while serum Metrnl level was measured by Enzyme-linked Immunosorbent Assay (ELISA) assay. According to the CIMT status, the subjects were divided into two groups: T2DM without atherosclerosis (DM-AS, CIMT<1.0mm) and T2DM with atherosclerosis (DM+AS, CIMT ≥1.0mm or carotid plaque).
Results: Serum Metrnl level was significantly increased in DM+AS group as compared to DM-AS group (0.77 ± 0.24 vs 0.51 ± 0.28 ng/ml, P <0.05). Binary logistic regression analysis showed that T2DM subjects in the fourth quartile of Metrnl levels demonstrated significantly higher risk of the presence of carotid atherosclerosis (P <0.05). The ROC curve used to identify the diagnostic accuracy of serum Metrnl level in the predication of T2DM subjects with carotid atherosclerosis revealed an area under the curve (AUC) of 0.742. The optimal cut-off value was 0.61 ng/ml with a sensitivity of 77.6% and a specificity of 58.8%.
Conclusions: This cross-sectional study clearly demonstrated that elevated serum Metrnl level was significantly associated with higher risk of the presence of carotid atherosclerosis. Metrnl is a promising therapeutic target for T2DM and its macro-vascular diseases.
{"title":"Association between Metrnl and carotid atherosclerosis in patients with type 2 diabetes mellitus.","authors":"Chunmei Li, Qiongzhu Huang, Yanqian Zhuang, Pin Chen, Yiyang Lin","doi":"10.3389/fendo.2024.1414508","DOIUrl":"https://doi.org/10.3389/fendo.2024.1414508","url":null,"abstract":"<p><strong>Objective: </strong>Type 2 diabetes mellitus (T2DM) is a major cause of atherosclerosis, as well as an independent risk factor of cardiovascular adverse events. We aimed to evaluate the association of serum Meteorin-like protein (Metrnl) level with carotid atherosclerosis as determined by carotid intima-media thickness (CIMT) status in subjects with T2DM.</p><p><strong>Methods: </strong>This cross-sectional study included 83 T2DM subjects without pre-existing cardiovascular diseases. CIMT was measured by color doppler ultrasonography, while serum Metrnl level was measured by Enzyme-linked Immunosorbent Assay (ELISA) assay. According to the CIMT status, the subjects were divided into two groups: T2DM without atherosclerosis (DM-AS, CIMT<1.0mm) and T2DM with atherosclerosis (DM+AS, CIMT ≥1.0mm or carotid plaque).</p><p><strong>Results: </strong>Serum Metrnl level was significantly increased in DM+AS group as compared to DM-AS group (0.77 ± 0.24 vs 0.51 ± 0.28 ng/ml, <i>P <</i>0.05). Binary logistic regression analysis showed that T2DM subjects in the fourth quartile of Metrnl levels demonstrated significantly higher risk of the presence of carotid atherosclerosis (<i>P <</i>0.05). The ROC curve used to identify the diagnostic accuracy of serum Metrnl level in the predication of T2DM subjects with carotid atherosclerosis revealed an area under the curve (AUC) of 0.742. The optimal cut-off value was 0.61 ng/ml with a sensitivity of 77.6% and a specificity of 58.8%.</p><p><strong>Conclusions: </strong>This cross-sectional study clearly demonstrated that elevated serum Metrnl level was significantly associated with higher risk of the presence of carotid atherosclerosis. Metrnl is a promising therapeutic target for T2DM and its macro-vascular diseases.</p>","PeriodicalId":12447,"journal":{"name":"Frontiers in Endocrinology","volume":"15 ","pages":"1414508"},"PeriodicalIF":3.9,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11750675/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143058426","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Bariatric surgery is an effective treatment for type 2 Diabetes Mellitus (T2DM), yet the precise mechanisms underlying its effectiveness remain incompletely understood. While previous research has emphasized the role of rearrangement of the gastrointestinal anatomy, gaps persist regarding the specific impact on the gut microbiota and barriers within the biliopancreatic, alimentary, and common limbs. This study aimed to investigate the effects of duodenal-jejunal bypass (DJB) surgery on obese T2DM mice. We performed DJB and SHAM surgery in obese T2DM mice to investigate changes in the gut microbiota and barrier across different intestinal limbs. The effects on serum metabolism and potential associations with T2DM improvement were also investigated. Following DJB surgery, there was an increased abundance of commensals across various limbs. Additionally, the surgery improved intestinal permeability and inflammation in the alimentary and common limbs, while reducing inflammation in the biliopancreatic limbs. Furthermore, DJB surgery also improved T2DM by increasing L-glutamine, short-chain fatty acids, and bile acids and decreasing branched-chain amino acids. This study underscores the role of intestinal rearrangement in reshaping gut microbiota composition and enhancing gut barrier function, thereby contributing to the amelioration of T2DM following bariatric surgery, and providing new insights for further research on bariatric surgery.
{"title":"Intestinal rearrangement of biliopancreatic limbs, alimentary limbs, and common limbs in obese type 2 diabetic mice after duodenal jejunal bypass surgery.","authors":"Heng Li, Jipei He, Jie Hou, Chengjun He, Xiaojiang Dai, Zhigao Song, Qing Liu, Zixin Wang, Hongyan Huang, Yunfa Ding, Tengfei Qi, Hongbin Zhang, Liangping Wu","doi":"10.3389/fendo.2024.1456885","DOIUrl":"10.3389/fendo.2024.1456885","url":null,"abstract":"<p><p>Bariatric surgery is an effective treatment for type 2 Diabetes Mellitus (T2DM), yet the precise mechanisms underlying its effectiveness remain incompletely understood. While previous research has emphasized the role of rearrangement of the gastrointestinal anatomy, gaps persist regarding the specific impact on the gut microbiota and barriers within the biliopancreatic, alimentary, and common limbs. This study aimed to investigate the effects of duodenal-jejunal bypass (DJB) surgery on obese T2DM mice. We performed DJB and SHAM surgery in obese T2DM mice to investigate changes in the gut microbiota and barrier across different intestinal limbs. The effects on serum metabolism and potential associations with T2DM improvement were also investigated. Following DJB surgery, there was an increased abundance of commensals across various limbs. Additionally, the surgery improved intestinal permeability and inflammation in the alimentary and common limbs, while reducing inflammation in the biliopancreatic limbs. Furthermore, DJB surgery also improved T2DM by increasing L-glutamine, short-chain fatty acids, and bile acids and decreasing branched-chain amino acids. This study underscores the role of intestinal rearrangement in reshaping gut microbiota composition and enhancing gut barrier function, thereby contributing to the amelioration of T2DM following bariatric surgery, and providing new insights for further research on bariatric surgery.</p>","PeriodicalId":12447,"journal":{"name":"Frontiers in Endocrinology","volume":"15 ","pages":"1456885"},"PeriodicalIF":3.9,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11750664/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143022534","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-07eCollection Date: 2024-01-01DOI: 10.3389/fendo.2024.1528968
Friederike Behler-Janbeck, Anke Baranowsky, Timur A Yorgan, Michelle Y Jaeckstein, Anna Worthmann, Marceline M Fuh, Karthikeyan Gunasekaran, Gisa Tiegs, Michael Amling, Thorsten Schinke, Joerg Heeren
[This corrects the article DOI: 10.3389/fendo.2024.1392418.].
[这更正了文章DOI: 10.3389/ fend.2024.1392418 .]。
{"title":"Corrigendum: The short-chain fatty acid receptors Gpr41/43 regulate bone mass by promoting adipogenic differentiation of mesenchymal stem cells.","authors":"Friederike Behler-Janbeck, Anke Baranowsky, Timur A Yorgan, Michelle Y Jaeckstein, Anna Worthmann, Marceline M Fuh, Karthikeyan Gunasekaran, Gisa Tiegs, Michael Amling, Thorsten Schinke, Joerg Heeren","doi":"10.3389/fendo.2024.1528968","DOIUrl":"https://doi.org/10.3389/fendo.2024.1528968","url":null,"abstract":"<p><p>[This corrects the article DOI: 10.3389/fendo.2024.1392418.].</p>","PeriodicalId":12447,"journal":{"name":"Frontiers in Endocrinology","volume":"15 ","pages":"1528968"},"PeriodicalIF":3.9,"publicationDate":"2025-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11748794/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143003080","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pancreatic intraepithelial neoplasia (PanIN) is the most common precursor lesion of pancreatic ductal adenocarcinoma (PDAC), which has poor prognosis with a short median overall survival of 6-12 months and a low 5-year survival rate of approximately 3%. It is crucial to remove PanIN lesions to prevent the development of invasive PDAC, as PDAC spreads rapidly outside the pancreas. This review aims to provide the latest knowledge on PanIN risk, pathology, cellular origin, genetic susceptibility, and diagnosis, while identifying research gaps that require further investigation in this understudied area of precancerous lesions. PanINs are classified into PanIN 1, PanIN 2, and PanIN 3, with PanIN 3 having the highest likelihood of developing into invasive PDAC. Differentiating between PanIN 2 and PanIN 3 is clinically significant. Genetic alterations found in PDAC are also present in PanIN and increase with the grade of PanIN. Imaging methods alone are insufficient for distinguishing PanIN, necessitating the use of genetic and molecular tests for identification. In addition, metabolomics technologies and miRNAs are playing an increasingly important role in the field of cancer diagnosis, offering more possibilities for efficient identification of PanIN. Although detecting and stratifying the risk of PanIN poses challenges, the combined utilization of imaging, genetics, and metabolomics holds promise for improving patient survival in this field.
{"title":"Identification and analysis of pancreatic intraepithelial neoplasia: opportunities and challenges.","authors":"Ling-Ling Pian, Mei-Hui Song, Teng-Fei Wang, Ling Qi, Tie-Li Peng, Ke-Ping Xie","doi":"10.3389/fendo.2024.1401829","DOIUrl":"10.3389/fendo.2024.1401829","url":null,"abstract":"<p><p>Pancreatic intraepithelial neoplasia (PanIN) is the most common precursor lesion of pancreatic ductal adenocarcinoma (PDAC), which has poor prognosis with a short median overall survival of 6-12 months and a low 5-year survival rate of approximately 3%. It is crucial to remove PanIN lesions to prevent the development of invasive PDAC, as PDAC spreads rapidly outside the pancreas. This review aims to provide the latest knowledge on PanIN risk, pathology, cellular origin, genetic susceptibility, and diagnosis, while identifying research gaps that require further investigation in this understudied area of precancerous lesions. PanINs are classified into PanIN 1, PanIN 2, and PanIN 3, with PanIN 3 having the highest likelihood of developing into invasive PDAC. Differentiating between PanIN 2 and PanIN 3 is clinically significant. Genetic alterations found in PDAC are also present in PanIN and increase with the grade of PanIN. Imaging methods alone are insufficient for distinguishing PanIN, necessitating the use of genetic and molecular tests for identification. In addition, metabolomics technologies and miRNAs are playing an increasingly important role in the field of cancer diagnosis, offering more possibilities for efficient identification of PanIN. Although detecting and stratifying the risk of PanIN poses challenges, the combined utilization of imaging, genetics, and metabolomics holds promise for improving patient survival in this field.</p>","PeriodicalId":12447,"journal":{"name":"Frontiers in Endocrinology","volume":"15 ","pages":"1401829"},"PeriodicalIF":3.9,"publicationDate":"2025-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11746065/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143003102","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}