1
|
Ma Y, Meng F, Lin Z, Chen Y, Lan T, Yang Z, Diao R, Zhang X, Chen Q, Zhang C, Tian Y, Li C, Fang W, Liang X, Zhang X. Bioengineering Platelets Presenting PD-L1, Galectin-9 and BTLA to Ameliorate Type 1 Diabetes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2501139. [PMID: 40019367 PMCID: PMC12021092 DOI: 10.1002/advs.202501139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 02/14/2025] [Indexed: 03/01/2025]
Abstract
Autoimmune destruction of pancreatic β-cells leads to impaired insulin production and onset of type 1 diabetes (T1D). Hence, immunomodulation of pancreas-infiltrated immune cells especially the β-cells autoreactive-T cells is a promising way to hinder and reverse the progress of T1D. Herein, megakaryocytes are primed with interferon-γ (IFN-γ) to produce platelets presenting high levels of immunosuppressive checkpoint ligands including programmed death-ligand 1 (PD-L1), Programmed Death-Ligand 2 (PD-L2), the B and T lymphocyte attenuator (BTLA) and Galectin-9 (Gal-9), termed as IFN-γ platelets. The IFN-γ platelets bound and interacted with T cells through immune checkpoint ligands and receptors, which efficaciously induced T cell exhaustion and apoptosis in vitro. Virtually, NOD diabetes mice received IFN-γ platelets treatments prominently preserved β-cell integrity and insulin production, ultimately hindering the progress to hyperglycemia. Intriguingly, both the amount and activity of the pancreas infiltrate-T cells intensively reduced, whereas the magnitude of regulatory T cells (Tregs) remarkably increased, which is attributed to IFN-γ platelets treatments. Moreover, IFN-γ platelets treatment instigated macrophage polarization toward an anti-inflammatory M2 phenotype that may stimulate pancreatic angiogenesis, and promote β-cell proliferation, consequently ameliorating the new-onset T1D.
Collapse
Grants
- 32371425 National Natural Science Foundation of China
- 32201084 National Natural Science Foundation of China
- JCYJ20240813151128037 Science, Technology & Innovation Commission of Shenzhen Municipality, Shenzhen Science and Technology Program
- RCYX20200714114643121 Science, Technology & Innovation Commission of Shenzhen Municipality, Shenzhen Science and Technology Program
- JCYJ20200109142610136 Science, Technology & Innovation Commission of Shenzhen Municipality, Shenzhen Science and Technology Program
- JCYJ20180507181654186 Science, Technology & Innovation Commission of Shenzhen Municipality, Shenzhen Science and Technology Program
- ZDSYS20220606100803007 Science, Technology & Innovation Commission of Shenzhen Municipality, Shenzhen Science and Technology Program
- 2022A1515012289 Natural Science Foundation of Guangdong Province
- GDMUB2022037 Doctoral personnel scientific research start-up Fund project of Guangdong Medical University
- 2024ZDZX2069 Key Field Special Programs of Guangdong Provincial Ordinary Colleges and Universities
- GDMULCJC2024114 Special Project for Clinical and Basic Sci & Tech Innovation of Guangdong Medical University
- National Natural Science Foundation of China
- Natural Science Foundation of Guangdong Province
Collapse
Affiliation(s)
- Yumeng Ma
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Fanqiang Meng
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Zhongda Lin
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Yanjun Chen
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Tianyu Lan
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Zhaoxin Yang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Rui Diao
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Xiaozhou Zhang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Qi Chen
- Guangdong Provincial Key Laboratory of Medical Molecular DiagnosticsKey Laboratory of Stem Cell and Regenerative Tissue EngineeringSchool of Basic Medical SciencesGuangdong Medical UniversityDongguan523808P. R. China
- The Affiliated Dongguan Songshan Lake Central HospitalGuangdong Medical UniversityDongguanGuangdong523806P. R. China
| | - Chi Zhang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Yishi Tian
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Chanjuan Li
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Wenli Fang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Xin Liang
- Guangdong Provincial Key Laboratory of Medical Molecular DiagnosticsKey Laboratory of Stem Cell and Regenerative Tissue EngineeringSchool of Basic Medical SciencesGuangdong Medical UniversityDongguan523808P. R. China
- The Affiliated Dongguan Songshan Lake Central HospitalGuangdong Medical UniversityDongguanGuangdong523806P. R. China
| | - Xudong Zhang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| |
Collapse
|
2
|
Wang Y, Bo Y, Liu Y, Zhou J, Nguyen D, Baskaran D, Liu Y, Wang H. Metabolic labeling and targeted modulation of adipocytes. Biomater Sci 2025; 13:434-445. [PMID: 39648977 PMCID: PMC11758917 DOI: 10.1039/d4bm01352b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
Adipocytes play a critical role in energy storage and endocrine signaling and are associated with various diseases such as cancer and diabetes. Facile strategies to engineer adipocytes have long been pursued for elucidating adipocyte biology and developing adipocyte-based therapies. Herein, we report metabolic glycan labeling of adipocytes and subsequent targeted modulation of adipocytes via click chemistry. We show that azido tags expressed on the surface of adipocytes can persist for over 4 days. By conjugating dibenzocyclooctyne (DBCO)-cargos onto azido-labeled adipocytes via click chemistry, the cargos can be retained on the adipocyte membrane for over 12 hours. We further show that signaling molecules including adiponectin, calreticulin, mannose-binding lectin 2, and milk fat globule-EGF factor 8 protein can be conjugated to adipocytes to orchestrate their phagocytosis by macrophages. The azido-labeled adipocytes grafted into mice can also mediate targeted conjugation of DBCO-cargos in vivo. This adipocyte labeling and targeting technology will facilitate the development of adipocyte-based therapies and provides a new platform for manipulating the interaction between adipocytes and other types of cells.
Collapse
Affiliation(s)
- Yueji Wang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Yang Bo
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Yusheng Liu
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Jiadiao Zhou
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Daniel Nguyen
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Dhyanesh Baskaran
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Yuan Liu
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Hua Wang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
- Cancer Center at Illinois (CCIL), Urbana, IL 61801, USA
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Carle College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Materials Research Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
3
|
Fu Q, Wu X, Lu Z, Chang Y, Jin Q, Jin T, Zhang M. TMEM205 induces TAM/M2 polarization to promote cisplatin resistance in gastric cancer. Gastric Cancer 2024; 27:998-1015. [PMID: 38850316 PMCID: PMC11335886 DOI: 10.1007/s10120-024-01517-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 05/26/2024] [Indexed: 06/10/2024]
Abstract
Cisplatin (DDP) is a basic chemotherapy drug for gastric cancer (GC). With the increase of DDP drug concentration in clinical treatment, cancer cells gradually became resistant. Therefore, it is necessary to find effective therapeutic targets to enhance the sensitivity of GC to DDP. Studies have shown that Transmembrane protein 205 (TMEM205) is overexpressed in DDP-resistant human epidermoid carcinoma cells and correlates with drug resistance, and database analyses show that TMEM 205 is also overexpressed in GC, but its role in cisplatin-resistant gastric cancer remains unclear. In this study, we chose a variety of experiments in vivo and vitro, aiming to investigate the role of TMEM 205 in cisplatin resistance in gastric cancer. The results showed that TMEM 205 promoted proliferation, stemness, epithelial-mesenchymal transition (EMT), migration and angiogenesis of gastric cancer cells through activation of the Wnt/β-catenin signaling pathway. In addition, TMEM205 promotes GC progression by inducing M2 polarization of tumor-associated macrophages (TAMs). These results suggest that TMEM205 may be an effective target to regulate the sensitivity of GC to DDP, providing a new therapeutic direction for clinical treatment.
Collapse
Affiliation(s)
- Qiang Fu
- Department of Ultrasound Medicine, Affiliated Hospital of Yanbian University, Yanji, 133000, Jilin, China
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji, 133002, China
- Key Laboratory of the Science and Technology Department of Jilin Province, Yanji, China
| | - Xuwei Wu
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji, 133002, China
- Key Laboratory of the Science and Technology Department of Jilin Province, Yanji, China
- Department of Pathology, Chifeng Municipal Hospital, Chifeng, 024000, China
| | - Zhongqi Lu
- Department of Ultrasound Medicine, Affiliated Hospital of Yanbian University, Yanji, 133000, Jilin, China
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji, 133002, China
- Key Laboratory of the Science and Technology Department of Jilin Province, Yanji, China
| | - Ying Chang
- Department of Ultrasound Medicine, Affiliated Hospital of Yanbian University, Yanji, 133000, Jilin, China
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji, 133002, China
- Key Laboratory of the Science and Technology Department of Jilin Province, Yanji, China
| | - Quanxin Jin
- Department of Immunology and Pathogenic Biology, Yanbian University Medical College, Yanji, China
| | - Tiefeng Jin
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji, 133002, China
- Key Laboratory of the Science and Technology Department of Jilin Province, Yanji, China
| | - Meihua Zhang
- Department of Health Examination Centre, Yanbian University Hospital, Yanji, 133002, China.
- Department of Ultrasound Medicine, Affiliated Hospital of Yanbian University, Yanji, 133000, Jilin, China.
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji, 133002, China.
| |
Collapse
|
4
|
Liu X, Tang Y, Luo Y, Gao Y, He L. Role and mechanism of specialized pro-resolving mediators in obesity-associated insulin resistance. Lipids Health Dis 2024; 23:234. [PMID: 39080624 PMCID: PMC11290132 DOI: 10.1186/s12944-024-02207-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 07/07/2024] [Indexed: 08/02/2024] Open
Abstract
With the changing times, obesity has become a characteristic epidemic in the context of the current era. Insulin resistance (IR) is most commonly caused by obesity, and IR is a common basis of the pathogenesis of many diseases such as cardiovascular disease, nonalcoholic fatty liver disease, and type 2 diabetes, which seriously threaten human life, as well as health. A major pathogenetic mechanism of obesity-associated IR has been found to be chronic low-grade inflammation in adipose tissue. Specialized pro-resolving mediators (SPMs) are novel lipid mediators that both function as "stop signals" for inflammatory reaction and promote inflammation to subside. In this article, we summarize the pathogenesis of obesity-associated IR and its treatments and outline the classification and biosynthesis of SPMs and their mechanisms and roles in the treatment of obesity-associated IR in order to explore the potential of SPMs for treating metabolic diseases linked with obesity-associated IR.
Collapse
Affiliation(s)
- Xinru Liu
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu Tang
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuanyuan Luo
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yongxiang Gao
- College of International Education, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Lisha He
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
5
|
Zhang Q, Chen R, Shi L, Zhao H, Yin F, Yu C, Wang Y, Lu P. Single-cell sequencing analysis of chronic subdural hematoma cell subpopulations and their potential therapeutic mechanisms. Brain Res Bull 2024; 211:110936. [PMID: 38554980 DOI: 10.1016/j.brainresbull.2024.110936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 03/19/2024] [Accepted: 03/27/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND Chronic subdural hematoma (CSDH) is a prevalent form of intracranial haemorrhage encountered in neurosurgical practice, and its incidence has notably risen in recent years. Currently, there is a lack of studies that have comprehensively classified the cells present in hematomas removed during surgery, and their correlation with CSDH recurrence remains elusive. This study aims to analyse the subcellular populations and occupancy levels within peripheral blood. METHODS This study analyses the subcellular populations and occupancy levels within peripheral blood and postoperatively removed hematomas by single-cell sequencing and attempts to analyse the effect of different cell occupancies within peripheral blood and intraoperatively removed hematomas on CSDH. RESULTS The single-cell sequencing results showed that the cells were classified into 25 clusters by differential gene and UMAP dimensionality reduction clustering analyses and further classified into 17 significant cell populations by cell markers: pDCs, CD8 T cells, CD4 T cells, MigDCs, cDC2s, cDC1s, plasma cells, neutrophils, naive B cells, NK cells, memory B cells, M2 macrophages, CD8 Teffs, CD8 MAIT cells, CD4 Tregs, CD19 B cells, and monocytes. Further research showed that the presence of more cDC2 and M2 macrophages recruited at the focal site in patients with CSDH and the upregulation of the level of T-cell occupancy may be a red flag for further brain damage. ROS, a marker of oxidative stress, was significantly upregulated in cDC2 cells and may mediate the functioning of transcription proteins of inflammatory factors, such as NFκB, which induced T cells' activation. Moreover, cDC2 may regulate M2 macrophage immune infiltration and anti-inflammatory activity by secreting IL1β and binding to M2 macrophage IL1R protein. CONCLUSION The detailed classification of cells in the peripheral blood and hematoma site of CSDH patients helps us to understand the mechanism of CSDH generation and the reduction in the probability of recurrence by regulating the ratio of cell subpopulations.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Neurosurgery, Sir Run Run Shaw Hospital Medical College, Zhejiang University, Hangzhou, Zhejiang Province 310016, China
| | - Rundong Chen
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Lufeng Shi
- Department of Neurosurgery, Sir Run Run Shaw Hospital Medical College, Zhejiang University, Hangzhou, Zhejiang Province 310016, China
| | - Hehe Zhao
- Department of Neurosurgery, Sir Run Run Shaw Hospital Medical College, Zhejiang University, Hangzhou, Zhejiang Province 310016, China
| | - Fei Yin
- Department of Neurosurgery, Sir Run Run Shaw Hospital Medical College, Zhejiang University, Hangzhou, Zhejiang Province 310016, China
| | - Cong Yu
- Department of Neurosurgery, Sir Run Run Shaw Hospital (Shaoxing), Shaoxing, Zhejiang Province 312300, China
| | - Yirong Wang
- Department of Neurosurgery, Sir Run Run Shaw Hospital Medical College, Zhejiang University, Hangzhou, Zhejiang Province 310016, China.
| | - Peng Lu
- Department of Neurosurgery, Sir Run Run Shaw Hospital Medical College, Zhejiang University, Hangzhou, Zhejiang Province 310016, China.
| |
Collapse
|
6
|
Hager M, Chang P, Lee M, Burns CM, Endicott SJ, Miller RA, Li X. Recapitulation of anti-aging phenotypes by global overexpression of PTEN in mice. GeroScience 2024; 46:2653-2670. [PMID: 38114855 PMCID: PMC10828233 DOI: 10.1007/s11357-023-01025-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 11/20/2023] [Indexed: 12/21/2023] Open
Abstract
The PTEN gene negatively regulates the oncogenic PI3K-AKT pathway by encoding a lipid and protein phosphatase that dephosphorylates lipid phosphatidylinositol-3,4,5-triphosphate (PIP3) resulting in the inhibition of PI3K and downstream inhibition of AKT. Overexpression of PTEN in mice leads to a longer lifespan compared to control littermates, although the mechanism is unknown. Here, we provide evidence that young adult PTENOE mice exhibit many characteristics shared by other slow-aging mouse models, including those with mutations that affect GH/IGF1 pathways, calorie-restricted mice, and mice treated with anti-aging drugs. PTENOE white adipose tissue (WAT) has increased UCP1, a protein linked to increased thermogenesis. WAT of PTENOE mice also shows a change in polarization of fat-associated macrophages, with elevated levels of arginase 1 (Arg1, characteristic of M2 macrophages) and decreased production of inducible nitric oxide synthase (iNOS, characteristic of M1 macrophages). Muscle and hippocampus showed increased expression of the myokine FNDC5, and higher levels of its cleavage product irisin in plasma, which has been linked to increased conversion of WAT to more thermogenic beige/brown adipose tissue. PTENOE mice also have an increase, in plasma and liver, of GPLD1, which is known to improve cognition in mice. Hippocampus of the PTENOE mice has elevation of both BDNF and DCX, indices of brain resilience and neurogenesis. These changes in fat, macrophages, liver, muscle, hippocampus, and plasma may be considered "aging rate indicators" in that they seem to be consistently changed across many of the long-lived mouse models and may help to extend lifespan by delaying many forms of late-life illness. Our new findings show that PTENOE mice can be added to the group of long-lived mice that share this multi-tissue suite of biochemical characteristics.
Collapse
Affiliation(s)
- Mary Hager
- College of Literature, Sciences, & the Arts, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Peter Chang
- College of Literature, Sciences, & the Arts, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Michael Lee
- College of Literature, Sciences, & the Arts, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Calvin M Burns
- Department of Pathology, University of Michigan School of Medicine, Room 3160, BSRB ,109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - S Joseph Endicott
- Department of Pathology, University of Michigan School of Medicine, Room 3160, BSRB ,109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
- University of Michigan Geriatrics Center, Ann Arbor, MI, 48109, USA
| | - Richard A Miller
- Department of Pathology, University of Michigan School of Medicine, Room 3160, BSRB ,109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
- University of Michigan Geriatrics Center, Ann Arbor, MI, 48109, USA
| | - Xinna Li
- Department of Pathology, University of Michigan School of Medicine, Room 3160, BSRB ,109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.
- University of Michigan Geriatrics Center, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
7
|
Valdes-Fernandez BN, Ruiz-Jimenez C, Armina-Rodriguez A, Mendez LB, Espino AM. Fasciola hepatica GST mu-class suppresses the cytokine storm induced by E. coli-lipopolysaccharide, whereas it modulates the dynamic of peritoneal macrophages in a mouse model and suppresses the classical activation of macrophages. Microbiol Spectr 2024; 12:e0347523. [PMID: 38018982 PMCID: PMC10782955 DOI: 10.1128/spectrum.03475-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 10/12/2023] [Indexed: 11/30/2023] Open
Abstract
IMPORTANCE Sepsis is the consequence of a systemic bacterial infection that exacerbates the immune cell's activation via bacterial products, resulting in the augmented release of inflammatory mediators. A critical factor in the pathogenesis of sepsis is the primary component of the outer membrane of Gram-negative bacteria known as lipopolysaccharide (LPS), which is sensed by TLR4. For this reason, scientists have aimed to develop antagonists able to block TLR4 and, thereby the cytokine storm. We report here that a mixture of mu-class isoforms from the F. hepatica GST protein family administered intraperitoneally 1 h prior to a lethal LPS injection can modulate the dynamics and abundance of large peritoneal macrophages in the peritoneal cavity of septic mice while significantly suppressing the LPS-induced cytokine storm in a mouse model of septic shock. These results suggest that native F. hepatica glutathione S-transferase is a promising candidate for drug development against endotoxemia and other inflammatory diseases.
Collapse
Affiliation(s)
| | | | | | - Loyda B. Mendez
- School of Sciences and Technologies, University Ana G. Mendez, Carolina, Puerto Rico
| | - Ana M. Espino
- Department of Microbiology, University of Puerto Rico, San Juan, Puerto Rico
| |
Collapse
|
8
|
Biagioli M, Marchianò S, Di Giorgio C, Bordoni M, Urbani G, Bellini R, Massa C, Sami Ullah Khan R, Roselli R, Chiara Monti M, Morretta E, Giordano A, Vellecco V, Bucci M, Jilani Iqbal A, Saviano A, Ab Mansour A, Ricci P, Distrutti E, Zampella A, Cieri E, Cirino G, Fiorucci S. Activation of GPBAR1 attenuates vascular inflammation and atherosclerosis in a mouse model of NAFLD-related cardiovascular disease. Biochem Pharmacol 2023; 218:115900. [PMID: 37926268 DOI: 10.1016/j.bcp.2023.115900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/16/2023] [Accepted: 10/26/2023] [Indexed: 11/07/2023]
Abstract
While patients with nonalcoholic fatty liver disease (NAFLD) are at increased risk to develop clinically meaningful cardiovascular diseases (CVD), there are no approved drug designed to target the liver and CVD component of NAFLD. GPBAR1, also known as TGR5, is a G protein coupled receptor for secondary bile acids. In this study we have investigated the effect of GPBAR1 activation by BAR501, a selective GPBAR1 agonist, in Apolipoprotein E deficient (ApoE-/-) mice fed a high fat diet and fructose (Western diet), a validated model of NAFLD-associated atherosclerosis. Using aortic samples from patients who underwent surgery for abdominal aneurism, and ex vivo experiments with endothelial cells and human macrophages, we were able to co-localize the expression of GPBAR1 in CD14+ and PECAM1+ cells. Similar findings were observed in the aortic plaques from ApoE-/- mice. Treating ApoE-/- mice with BAR501, 30 mg/kg for 14 weeks, attenuated the body weight gain while ameliorated the insulin sensitivity by increasing the plasma concentrations of GLP-1 and FGF15. Activation of GPBAR1 reduced the aorta thickness and severity of atherosclerotic lesions and decreased the amount of plaques macrophages. Treating ApoE-/- mice reshaped the aortic transcriptome promoting the expression of anti-inflammatory genes, including IL-10, as also confirmed by tSNE analysis of spleen-derived macrophages. Feeding ApoE-/- mice with BAR501 redirected the bile acid synthesis and the composition of the intestinal microbiota. In conclusion, GPBAR1 agonism attenuates systemic inflammation and improve metabolic profile in a genetic/dietetic model of atherosclerosis. BAR501 might be of utility in the treatment for NAFLD-related CVD.
Collapse
Affiliation(s)
- Michele Biagioli
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Silvia Marchianò
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Martina Bordoni
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Ginevra Urbani
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Rachele Bellini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Carmen Massa
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Rosalinda Roselli
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | | | - Elva Morretta
- Department of Pharmacy, University of Salerno, Salerno, Italy
| | - Antonino Giordano
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | | | - Asif Jilani Iqbal
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Anella Saviano
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Adel Ab Mansour
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Patrizia Ricci
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Angela Zampella
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Enrico Cieri
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Giuseppe Cirino
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Stefano Fiorucci
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy.
| |
Collapse
|
9
|
Zhang Y, Luo Y, Liu X, Kiupel M, Li A, Wang H, Mi QS, Xiao H. NCOA5 Haploinsufficiency in Myeloid-Lineage Cells Sufficiently Causes Nonalcoholic Steatohepatitis and Hepatocellular Carcinoma. Cell Mol Gastroenterol Hepatol 2023; 17:1-27. [PMID: 37734594 PMCID: PMC10665956 DOI: 10.1016/j.jcmgh.2023.09.007] [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: 07/20/2022] [Revised: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 09/23/2023]
Abstract
BACKGROUND & AIMS The nuclear receptor coactivator 5 (NCOA5) is a putative type 2 diabetes susceptibility gene. NCOA5 haploinsufficiency results in the spontaneous development of nonalcoholic fatty liver disease (NAFLD), insulin resistance, and hepatocellular carcinoma (HCC) in male mice; however, the cell-specific effect of NCOA5 haploinsufficiency in various types of cells, including macrophages, on the development of NAFLD and HCC remains unknown. METHODS Control and myeloid-lineage-specific Ncoa5 deletion (Ncoa5ΔM/+) mice fed a normal diet were examined for the development of NAFLD, nonalcoholic steatohepatitis (NASH), and HCC. Altered genes and signaling pathways in the intrahepatic macrophages of Ncoa5ΔM/+ male mice were analyzed and compared with those of obese human individuals. The role of platelet factor 4 (PF4) in macrophages and the underlying mechanism by which PF4 affects NAFLD/NASH were explored in vitro and in vivo. PF4 expression in HCC patient specimens and prognosis was examined. RESULTS Myeloid-lineage-specific Ncoa5 deletion sufficiently causes spontaneous NASH and HCC development in male mice fed a normal diet. PF4 overexpression in Ncoa5ΔM/+ intrahepatic macrophages is identified as a potent mediator to trigger lipid accumulation in hepatocytes by inducing lipogenesis-promoting gene expression. The transcriptome of intrahepatic macrophages from Ncoa5ΔM/+ male mice resembles that of obese human individuals. High PF4 expression correlated with poor prognosis of HCC patients and increased infiltrations of M2 macrophages, regulatory T cells, and myeloid-derived suppressor cells in HCCs. CONCLUSIONS Our findings reveal a novel mechanism for the onset of NAFLD/NASH and HCC initiated by NCOA5-deficient macrophages, suggesting the NCOA5-PF4 axis in macrophages as a potential target for developing preventive and therapeutic interventions against NAFLD/NASH and HCC.
Collapse
Affiliation(s)
- Yueqi Zhang
- Cell and Molecular Biology Program, Michigan State University, East Lansing, Michigan; Department of Physiology, Michigan State University, East Lansing, Michigan
| | - Yue Luo
- Department of Physiology, Michigan State University, East Lansing, Michigan; Cancer Center, Southern Medical University, Guangzhou, Guangdong, China; Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Xinhui Liu
- Department of Physiology, Michigan State University, East Lansing, Michigan; Cancer Center, Southern Medical University, Guangzhou, Guangdong, China; Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Matti Kiupel
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan
| | - Aimin Li
- Cancer Center, Southern Medical University, Guangzhou, Guangdong, China; Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Hongbing Wang
- Department of Physiology, Michigan State University, East Lansing, Michigan
| | - Qing-Sheng Mi
- Immunology Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, Michigan; Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health, Detroit, Michigan
| | - Hua Xiao
- Department of Physiology, Michigan State University, East Lansing, Michigan.
| |
Collapse
|
10
|
Kiseleva V, Vishnyakova P, Elchaninov A, Fatkhudinov T, Sukhikh G. Biochemical and molecular inducers and modulators of M2 macrophage polarization in clinical perspective. Int Immunopharmacol 2023; 122:110583. [PMID: 37423155 DOI: 10.1016/j.intimp.2023.110583] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/24/2023] [Accepted: 06/26/2023] [Indexed: 07/11/2023]
Abstract
Macrophages as innate immune cells with great plasticity are of great interest for cell therapy. There are two main macrophage populations - pro- and anti-inflammatory cells also known as M1 and M2. High potential in cancer research contributed to the in-depth study of the molecular processes leading to the polarization of macrophages into the M1 phenotype, and much less attention has been paid to anti-inflammatory M2 macrophages, which can be successfully used in cell therapy of inflammatory diseases. This review describes ontogenesis of macrophages, main functions of pro- and and-inflammatory cells and four M2 subpopulations characterized by different functionalities. Data on agents (cytokines, microRNAs, drugs, plant extracts) that may induce M2 polarization through the changes in microenvironment, metabolism, and efferocytosis are summarized. Finally, recent attempts at stable macrophage polarization using genetic modifications are described. This review may be helpful for researchers concerned with the problem of M2 macrophage polarization and potential use of these anti-inflammatory cells for the purposes of regenerative medicine.
Collapse
Affiliation(s)
- Viktoriia Kiseleva
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia; Peoples' Friendship University of Russia, Moscow, Russia.
| | - Polina Vishnyakova
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia; Peoples' Friendship University of Russia, Moscow, Russia
| | - Andrey Elchaninov
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia; Peoples' Friendship University of Russia, Moscow, Russia; Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution "Petrovsky National Research Centre of Surgery", Moscow, Russia
| | - Timur Fatkhudinov
- Peoples' Friendship University of Russia, Moscow, Russia; Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution "Petrovsky National Research Centre of Surgery", Moscow, Russia
| | - Gennady Sukhikh
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| |
Collapse
|
11
|
Li X, McPherson M, Hager M, Lee M, Chang P, Miller RA. Four anti-aging drugs and calorie-restricted diet produce parallel effects in fat, brain, muscle, macrophages, and plasma of young mice. GeroScience 2023; 45:2495-2510. [PMID: 36920743 PMCID: PMC10651632 DOI: 10.1007/s11357-023-00770-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 03/06/2023] [Indexed: 03/16/2023] Open
Abstract
Average and maximal lifespan can be increased in mice, in one or both sexes, by four drugs: rapamycin, acarbose, 17a-estradiol, and canagliflozin. We show here that these four drugs, as well as a calorie-restricted diet, can induce a common set of changes in fat, macrophages, plasma, muscle, and brain when evaluated in young adults at 12 months of age. These shared traits include an increase in uncoupling protein UCP1 in brown fat and in subcutaneous and intra-abdominal white fat, a decline in proinflammatory M1 macrophages and corresponding increase in anti-inflammatory M2 macrophages, an increase in muscle fibronectin type III domain containing 5 (FNDC5) and its cleavage product irisin, and higher levels of doublecortin (DCX) and brain-derived neurotrophic factor (BDNF) in brain. Each of these proteins is thought to play a role in one or more age-related diseases, including metabolic, inflammatory, and neurodegenerative diseases. We have previously shown that the same suite of changes is seen in each of four varieties of slow-aging single-gene mutant mice. We propose that these changes may be a part of a shared common pathway that is seen in slow-aging mice whether the delayed aging is due to a mutation, a low-calorie diet, or a drug.
Collapse
Affiliation(s)
- Xinna Li
- Department of Pathology, University of Michigan School of Medicine, BSRB, 109 Zina Pitcher Place, RoomAnn Arbor, MI, 316048109-2200, USA.
| | - Madaline McPherson
- College of Literature, Science, & the Arts, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Mary Hager
- College of Literature, Science, & the Arts, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Michael Lee
- College of Literature, Science, & the Arts, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Peter Chang
- College of Literature, Science, & the Arts, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Richard A Miller
- Department of Pathology, University of Michigan School of Medicine, BSRB, 109 Zina Pitcher Place, RoomAnn Arbor, MI, 316048109-2200, USA
- University of Michigan Geriatrics Center, Ann Arbor, MI, 48109, USA
| |
Collapse
|
12
|
Li X, Hager M, McPherson M, Lee M, Hagalwadi R, Skinner ME, Lombard D, Miller RA. Recapitulation of anti-aging phenotypes by global, but not by muscle-specific, deletion of PAPP-A in mice. GeroScience 2023; 45:931-948. [PMID: 36542300 PMCID: PMC9886707 DOI: 10.1007/s11357-022-00692-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 11/15/2022] [Indexed: 12/24/2022] Open
Abstract
Deletion of pregnancy-associated plasma protein-A (PAPP-A), a protease that cleaves some but not all IGF1 binding proteins, postpones late-life diseases and extends lifespan in mice, but the mechanism of this effect is unknown. Here we show that PAPP-A knockout (PKO) mice display a set of changes, in multiple tissues, that are characteristic of other varieties of slow-aging mice with alterations in GH production or GH responsiveness, including Ames dwarf, Snell dwarf, and GHRKO mice. PKO mice have elevated UCP1 in brown and white adipose tissues (WAT), and a change in fat-associated macrophage subsets that leads to diminished production of inflammatory cytokines. PKO mice also show increased levels of muscle FNDC5 and its cleavage product, the myokine irisin, thought to cause changes in fat cell differentiation. PKO mice have elevated production of hepatic GPLD1 and plasma GPLD1, consistent with their elevation of hippocampal BDNF and DCX, used as indices of neurogenesis. In contrast, disruption of PAPP-A limited to muscle ("muPKO" mice) produces an unexpectedly complex set of changes, in most cases opposite in direction from those seen in PKO mice. These include declines in WAT UCP1, increases in inflammatory macrophages and cytokines in WAT, and a decline in muscle FNDC5 and plasma irisin. muPKO mice do, however, resemble global PKO mice in their elevation of hippocampal BDNF and DCX. The data for the PKO mice support the idea that these changes in fat, macrophages, liver, muscle, plasma, and brain are consistent and biologically significant features of the slow-aging phenotype in mice. The results on the muPKO mice provide a foundation for further investigation of the complex, local, and global circuits by which PAPP-A modulates signals ordinarily controlled by GH and/or IGF1.
Collapse
Affiliation(s)
- Xinna Li
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, 48109, USA.
- , Ann Arbor, USA.
| | - Mary Hager
- College of Literature, Sciences, & the Arts, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Madaline McPherson
- College of Literature, Sciences, & the Arts, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Michael Lee
- College of Literature, Sciences, & the Arts, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Riha Hagalwadi
- College of Literature, Sciences, & the Arts, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Mary E Skinner
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, 48109, USA
| | - David Lombard
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, 48109, USA
- Sylvester Cancer Center, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Richard A Miller
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, 48109, USA
- University of Michigan Geriatrics Center, Ann Arbor, MI, 48109, USA
| |
Collapse
|
13
|
Röszer T. Metabolic impact of adipose tissue macrophages in the early postnatal life. J Leukoc Biol 2022; 112:1515-1524. [PMID: 35899927 PMCID: PMC9796690 DOI: 10.1002/jlb.3mr0722-201r] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 07/02/2022] [Indexed: 01/07/2023] Open
Abstract
Adipose tissue macrophages (ATMs) play key roles in metabolic inflammation, insulin resistance, adipose tissue fibrosis, and immune disorders associated with obesity. Research on ATM biology has mostly been conducted in the setting of adult obesity, since adipocyte hypertrophy is associated with a significant increase in ATM number. Signals that control ATM activation toward a proinflammatory or a proresolving phenotype also determine the developmental program and lipid metabolism of adipocytes after birth. ATMs are present at birth and actively participate in the synthesis of mediators, which induce lipolysis, mitobiogenesis, and mitochondrial uncoupling in adipocytes. ATMs in the newborn and the infant promote a lipolytic and fatty acid oxidizing adipocyte phenotype, which is essential to support the lipid-fueled metabolism, to maintain nonshivering thermogenesis and counteract an excessive adipose tissue expansion. Since adipose tissue metabolism in the early postnatal life determines obesity status in adulthood, early-life ATM functions may have a life-long impact.
Collapse
Affiliation(s)
- Tamás Röszer
- Division of Pediatric Obesity, Children's Hospital and Institute of PediatricsUniversity of DebrecenDebrecenHungary,Institute of NeurobiologyUlm UniversityUlmGermany
| |
Collapse
|
14
|
Laursen TL, Mellemkjær A, Møller HJ, Grønbæk H, Kazankov K. Spotlight on liver macrophages for halting injury and progression in non-alcoholic fatty liver disease. Expert Opin Ther Targets 2022; 26:697-705. [PMID: 36205054 DOI: 10.1080/14728222.2022.2132145] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
INTRODUCTION Non-alcoholic fatty liver disease (NAFLD) is considered the hepatic manifestation of the metabolic syndrome and is rapidly emerging as the leading cause of liver-related morbidity and mortality. Macrophages play an essential role in the development and progression of NAFLD. AREAS COVERED In this review, we provide an update on recent studies of drugs, which directly or indirectly affect macrophages in NAFLD, and discuss the implication of macrophage biomarkers to monitor the disease stage and progression/regression. EXPERT OPINION There is an unmet need for better understanding of disease pathogenesis from hepatic fat accumulation to disease progression with inflammation and fibrosis. We expect that future research will uncover additional objects/pathways as treatment targets. We speculate that this will involve better characterization of the gut microbiome, damage-associated molecular patterns (DAMPS) or molecules and pathways involved in development of DAMPS, and advanced molecular biology studies including single-cell sequencing of macrophage subpopulations. In addition, we speculate that studies focusing on pharmaceuticals that improve insulin resistance, diminish the metabolic syndrome and reduce fibrosis will prevail.
Collapse
Affiliation(s)
- Tea Lund Laursen
- Department of Medicine, Randers Regional Hospital, Randers, Denmark
| | - Anders Mellemkjær
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| | - Holger Jon Møller
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Henning Grønbæk
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| | - Konstantin Kazankov
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
15
|
Moritani Y, Hasegawa T, Yamamoto T, Hongo H, Yimin, Abe M, Yoshino H, Nakanishi K, Maruoka H, Ishizu H, Shimizu T, Takahata M, Iwasaki N, Li M, Tei K, Ohiro Y, Amizuka N. Histochemical assessment of accelerated bone remodeling and reduced mineralization in Il-6 deficient mice. J Oral Biosci 2022; 64:410-421. [DOI: 10.1016/j.job.2022.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/05/2022] [Accepted: 10/06/2022] [Indexed: 11/05/2022]
|
16
|
Al Madhoun A, Kochumon S, Al-Rashed F, Sindhu S, Thomas R, Miranda L, Al-Mulla F, Ahmad R. Dectin-1 as a Potential Inflammatory Biomarker for Metabolic Inflammation in Adipose Tissue of Individuals with Obesity. Cells 2022; 11:2879. [PMID: 36139454 PMCID: PMC9496833 DOI: 10.3390/cells11182879] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/02/2022] [Accepted: 09/10/2022] [Indexed: 11/17/2022] Open
Abstract
In obesity, macrophage activation and infiltration in adipose tissue (AT) underlie chronic low-grade inflammation-induced insulin resistance. Although dectin-1 is primarily a pathogen recognition receptor and innate immune response modulator, its role in metabolic syndromes remains to be clarified. This study aimed to investigate the dectin-1 gene expression in subcutaneous AT in the context of obesity and associated inflammatory markers. Subcutaneous AT biopsies were collected from 59 nondiabetic (lean/overweight/obese) individuals. AT gene expression levels of dectin-1 and inflammatory markers were determined via real-time reverse transcriptase-quantitative polymerase chain reaction. Dectin-1 protein expression was assessed using immunohistochemistry. Plasma lipid profiles were measured by ELISA. AT dectin-1 transcripts and proteins were significantly elevated in obese as compared to lean individuals. AT dectin-1 transcripts correlated positively with body mass index and fat percentage (r ≥ 0.340, p ≤ 0.017). AT dectin-1 RNA levels correlated positively with clinical parameters, including plasma C-reactive protein and CCL5/RANTES, but negatively with that of adiponectin. The expression of dectin-1 transcripts was associated with that of various proinflammatory cytokines, chemokines, and their cognate receptors (r ≥ 0.300, p ≤ 0.05), but not with anti-inflammatory markers. Dectin-1 and members of the TLR signaling cascade were found to be significantly associated, suggesting an interplay between the two pathways. Dectin-1 expression was correlated with monocyte/macrophage markers, including CD16, CD68, CD86, and CD163, suggesting its monocytes/macrophage association in an adipose inflammatory microenvironment. Dectin-1 expression was independently predicted by CCR5, CCL20, TLR2, and MyD88. In conclusion, dectin-1 may be regarded as an AT biomarker of metabolic inflammation in obesity.
Collapse
Affiliation(s)
- Ashraf Al Madhoun
- Animal and Imaging Core Facilities, Dasman Diabetes Institute, Dasman 15462, Kuwait
- Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Shihab Kochumon
- Immunology and Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Fatema Al-Rashed
- Immunology and Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Sardar Sindhu
- Animal and Imaging Core Facilities, Dasman Diabetes Institute, Dasman 15462, Kuwait
- Immunology and Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Reeby Thomas
- Immunology and Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Lavina Miranda
- Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Fahd Al-Mulla
- Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Rasheed Ahmad
- Immunology and Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait
| |
Collapse
|
17
|
Fiorucci S, Zampella A, Ricci P, Distrutti E, Biagioli M. Immunomodulatory functions of FXR. Mol Cell Endocrinol 2022; 551:111650. [PMID: 35472625 DOI: 10.1016/j.mce.2022.111650] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/31/2022] [Accepted: 04/04/2022] [Indexed: 02/08/2023]
Abstract
The Farnesoid-x-receptor (FXR) is a bile acids sensor activated in humans by primary bile acids. FXR is mostly expressed in liver, intestine and adrenal glands but also by cells of innate immunity, including macrophages, liver resident macrophages, the Kupffer cells, natural killer cells and dendritic cells. In normal physiology and clinical disorders, cells of innate immunity mediate communications between liver, intestine and adipose tissues. In addition to FXR, the G protein coupled receptor (GPBAR1), that is mainly activated by secondary bile acids, whose expression largely overlaps FXR, modulates chemical communications from the intestinal microbiota and the host's immune system, integrating epithelial cells and immune cells in the entero-hepatic system, providing a mechanism for development of a tolerogenic state toward the intestinal microbiota. Disruption of FXR results in generalized inflammation and disrupted bile acids metabolism. While FXR agonism in preclinical models provides counter-regulatory signals that attenuate inflammation-driven immune dysfunction in a variety of liver and intestinal disease models, the clinical relevance of these mechanisms in the setting of FXR-related disorders remain poorly defined.
Collapse
Affiliation(s)
- Stefano Fiorucci
- Dipartimento di Medicina e Chirurgia, Università di Perugia, Perugia, Italy. http://www.gastroenterologia.unipg.it
| | - Angela Zampella
- University of Naples Federico II, Department of Pharmacy, Naples, Italy
| | - Patrizia Ricci
- Dipartimento di Medicina e Chirurgia, Università di Perugia, Perugia, Italy
| | - Eleonora Distrutti
- SC di Gastroenterologia ed Epatologia, Azienda Ospedaliera di Perugia, Perugia, Italy
| | - Michele Biagioli
- Dipartimento di Medicina e Chirurgia, Università di Perugia, Perugia, Italy
| |
Collapse
|
18
|
Yakupova EI, Maleev GV, Krivtsov AV, Plotnikov EY. Macrophage polarization in hypoxia and ischemia/reperfusion: Insights into the role of energetic metabolism. Exp Biol Med (Maywood) 2022; 247:958-971. [PMID: 35220781 PMCID: PMC9189569 DOI: 10.1177/15353702221080130] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2023] Open
Abstract
Macrophages, the key cells of innate immunity, possess wide phenotypical and functional heterogeneity. In vitro studies showed that microenvironment signals could induce the so-called polarization of macrophages into two phenotypes: classically activated macrophages (M1) or alternatively activated macrophages (M2). Functionally, they are considered as proinflammatory and anti-inflammatory/pro-regenerative, respectively. However, in vivo studies into macrophage states revealed a continuum of phenotypes from M1 to M2 state instead of the clearly distinguished extreme phenotypes. An important role in determining the type of polarization of macrophages is played by energy metabolism, including the activity of oxidative phosphorylation. In this regard, hypoxia and ischemia that affect cellular energetics can modulate macrophage polarization. Here, we overview the data on macrophage polarization during metabolic shift-associated pathologies including ischemia and ischemia/reperfusion in various organs and discuss the role of energy metabolism potentially triggering the macrophage polarization.
Collapse
Affiliation(s)
- Elmira I Yakupova
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119234, Russia
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino 142290, Russia
| | - Grigoriy V Maleev
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka 142432, Russia
| | - Andrei V Krivtsov
- Center for Pediatric Cancer Therapeutics, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Egor Y Plotnikov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119234, Russia
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Moscow 117997, Russia
| |
Collapse
|
19
|
Harasgama JC, Kasthuriarachchi TDW, Sirisena DMKP, Kwon H, Lee S, Wan Q, Lee J. Modulation of fish immune response by interferon regulatory factor 4 in redlip mullet (Liza haematocheilus): Delineation through expression profiling, antiviral assay, and macrophage polarization analysis. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 130:104356. [PMID: 35065138 DOI: 10.1016/j.dci.2022.104356] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 01/17/2022] [Accepted: 01/17/2022] [Indexed: 06/14/2023]
Abstract
Interferon regulatory factor 4 (IRF4) is a crucial member of IRF family, which acts as an imperative transcription factor in the development and maturation of multiple lineages of blood cells and also plays a pivotal role in host defense against microbial infections. In the present study, we aimed to investigate the detailed structural and functional aspects of a redlip mullet IRF4 homolog (LhIRF4). The LhIRF4 open reading frame consists of 1347 base pairs encoding 449 amino acids, with the DNA-binding domain sharing significant homology with that of other vertebrate IRF4 homologs. The highest transcription levels of LhIRF4 were observed in the mullet intestine and spleen under normal physiological conditions. Furthermore, a time-dependent upregulation of LhIRF4 transcription was observed in the spleen and head kidney tissues upon pathogenic challenges. When overexpressed in mullet cells, LhIRF4 was localized to the nucleus and significantly stimulated the transcription of several host antiviral genes. Moreover, the overexpression of LhIRF4 strongly inhibited the replication of viral hemorrhagic septicemia virus (VHSV) in vitro. The function of LhIRF4 in regulation of macrophage M2 polarization has also been evidently demonstrated in RAW 264.7 cells. Taken together, our findings indicate the profound role of LhIRF4 in modulating immune responses against microbial infections in redlip mullet.
Collapse
Affiliation(s)
- J C Harasgama
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province, 63333, Republic of Korea
| | - T D W Kasthuriarachchi
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province, 63333, Republic of Korea
| | - D M K P Sirisena
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province, 63333, Republic of Korea
| | - Hyukjae Kwon
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province, 63333, Republic of Korea
| | - Seongdo Lee
- General Affairs Division, National Fishery Products Quality Management Service, Busan, 49111, Republic of Korea
| | - Qiang Wan
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province, 63333, Republic of Korea.
| | - Jehee Lee
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province, 63333, Republic of Korea.
| |
Collapse
|
20
|
Kwack K, Zhang L, Sohn J, Maglaras V, Thiyagarajan R, Kirkwood K. Novel Preosteoclast Populations in Obesity-Associated Periodontal Disease. J Dent Res 2022; 101:348-356. [PMID: 34636272 PMCID: PMC8982008 DOI: 10.1177/00220345211040729] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Although there is a clear relationship between the degree of obesity and periodontal disease incidence, the mechanisms that underpin the links between these conditions are not completely understood. Understanding that myeloid-derived suppressor cells (MDSCs) are expanded during obesity and operate in a context-defined manner, we addressed the potential role of MDSCs to contribute toward obesity-associated periodontal disease. Flow cytometry revealed that in the spleen of mice fed a high-fat diet (HFD), expansion in monocytic MDSCs (M-MDSCs) significantly increased when compared with mice fed a low-fat diet (LFD). In the osteoclast differentiation assay, M-MDSCs isolated from the bone marrow of HFD-fed mice showed a larger number and area of osteoclasts with a greater number of nuclei. In the M-MDSCs of HFD-fed mice, several osteoclast-related genes were significantly elevated when compared with LFD-fed mice according to a focused transcriptomic platform. In experimental periodontitis, the number and percentage of M-MDSCs were greater, with a significantly larger increase in HFD-fed mice versus LFD-fed mice. In the spleen, the percentage of M-MDSCs was significantly higher in HFD-fed periodontitis-induced (PI) mice than in LFD-PI mice. Alveolar bone volume fraction was significantly reduced in experimental periodontitis and was further decreased in HFD-PI mice as compared with LFD-PI mice. The inflammation score was significantly higher in HFD-PI mice versus LFD-PI mice, with a concomitant increase in TRAP staining for osteoclast number and area in HFD-PI mice over LFD-PI mice. These data support the concept that M-MDSC expansion during obesity to become osteoclasts during periodontitis is related to increased alveolar bone destruction, providing a more detailed mechanistic appreciation of the interconnection between obesity and periodontitis.
Collapse
Affiliation(s)
- K.H. Kwack
- Department of Oral Biology,
University at Buffalo, Buffalo, NY, USA
| | - L. Zhang
- Department of Oral Biology,
University at Buffalo, Buffalo, NY, USA
| | - J. Sohn
- Department of Oral Biology,
University at Buffalo, Buffalo, NY, USA,Department of Medicine,
University at Buffalo, Buffalo, NY, USA,Department of Genetics, Genomics,
and Bioinformatics Program, University at Buffalo, Buffalo, NY, USA
| | - V. Maglaras
- Department of Oral Biology,
University at Buffalo, Buffalo, NY, USA
| | - R. Thiyagarajan
- Department of Medicine,
University at Buffalo, Buffalo, NY, USA,Research Service, Western New
York Veterans Affairs Healthcare Service, Buffalo, NY, USA
| | - K.L. Kirkwood
- Department of Oral Biology,
University at Buffalo, Buffalo, NY, USA,Department of Head and
Neck/Plastic and Reconstructive Surgery, Roswell Park Comprehensive Cancer
Center, Buffalo, NY, USA,K.L. Kirkwood, Department of Oral
Biology, School of Dental Medicine, University at Buffalo, The State
University of New York, 645 Biomedical Research Building, 3435 Main
St, Buffalo, NY 14214-8006, USA.
| |
Collapse
|
21
|
The Preliminary Evaluation of Epigenetic Modifications Regulating the Expression of IL10 in Insulin-Resistant Adipocytes. Genes (Basel) 2022; 13:genes13020294. [PMID: 35205339 PMCID: PMC8872567 DOI: 10.3390/genes13020294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/28/2022] [Accepted: 01/30/2022] [Indexed: 11/16/2022] Open
Abstract
A higher level of IL10 expression in obesity and insulin resistance was observed in both human and mouse WAT. In our research, we analyzed the influence of insulin resistance on epigenetic modification within the promoter region IL10 gene and the potential influence of these modifications on its expression. Studies were performed using two cell models for the analysis: human, preadipocytes derived from adipose (visceral and subcutaneous) tissues and murine 3T3-L1 fibroblasts. We demonstrated a significant increase in the IL10 expression level, IL10 promoter region methylation, and histone 3 epigenetic modifications: H3K4me and H3K9/14ac, in insulin resistance cells (IR) from SAT cell culture. In IR cells from VAT cell culture, we observed decreased IL10 expression with a simultaneous increase of IL10 promoter region methylation. In IR cells from 3T3L1 cell culture, we observed the increased expression of IL10 as well as the decreased levels of methylation in the IL10 promoter region and histone methylation (H3K4me) and acetylation (H3K9/14ac). The presented analyses suggest a potential impact of epigenetic modifications on gene expression and a potential mutual influence of epigenetic modifications on each other or the activation of specific epigenetic regulation at a different stage of the development of insulin resistance in cells.
Collapse
|
22
|
Hastreiter AA, Paredes LC, Saraiva Camara NO. Metabolic requirement for macrophages. MACROPHAGES IN THE HUMAN BODY 2022:49-66. [DOI: 10.1016/b978-0-12-821385-8.00010-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
23
|
Goswami KK, Bose A, Baral R. Macrophages in tumor: An inflammatory perspective. Clin Immunol 2021; 232:108875. [PMID: 34740843 DOI: 10.1016/j.clim.2021.108875] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 10/07/2021] [Accepted: 10/27/2021] [Indexed: 01/08/2023]
Abstract
Inflammation is a part of carefully co-ordinated healing immune exercise to eliminate injurious stimuli. However, in substantial number of cancer types, it contributes in shaping up of robust tumor microenvironment (TME). Solid TME promotes infiltration of tumor associated macrophages (TAMs) that contributes to cancer promotion. TAMs are functionally heterogeneous and display an extraordinary degree of plasticity, which allow 'Switching' of macrophages into an 'M2', phenotype, linked with immunosuppression, advancement of tumor angiogenesis with metastatic consequences. In contrary to the classical M1 macrophages, these M2 TAMs are high-IL-10, TGF-β secreting-'anti-inflammatory'. In this review, we will discuss the modes of infiltration and switching of TAMs into M2 anti-inflammatory state in the TME to promote immunosuppression and inflammation-driven cancer.
Collapse
Affiliation(s)
- Kuntal Kanti Goswami
- Department of Microbiology, Asutosh College, 92, S. P. Mukherjee Road, Kolkata 700026, India.
| | - Anamika Bose
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India
| | - Rathindranath Baral
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India
| |
Collapse
|
24
|
The Role of Obesity-Induced Perivascular Adipose Tissue (PVAT) Dysfunction in Vascular Homeostasis. Nutrients 2021; 13:nu13113843. [PMID: 34836100 PMCID: PMC8621306 DOI: 10.3390/nu13113843] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 10/23/2021] [Accepted: 10/26/2021] [Indexed: 12/17/2022] Open
Abstract
Perivascular adipose tissue (PVAT) is an additional special type of adipose tissue surrounding blood vessels. Under physiological conditions, PVAT plays a significant role in regulation of vascular tone, intravascular thermoregulation, and vascular smooth muscle cell (VSMC) proliferation. PVAT is responsible for releasing adipocytes-derived relaxing factors (ADRF) and perivascular-derived relaxing factors (PDRF), which have anticontractile properties. Obesity induces increased oxidative stress, an inflammatory state, and hypoxia, which contribute to PVAT dysfunction. The exact mechanism of vascular dysfunction in obesity is still not well clarified; however, there are some pathways such as renin-angiotensin-aldosterone system (RAAS) disorders and PVAT-derived factor dysregulation, which are involved in hypertension and endothelial dysfunction development. Physical activity has a beneficial effect on PVAT function among obese patients by reducing the oxidative stress and inflammatory state. Diet, which is the second most beneficial non-invasive strategy in obesity treatment, may have a positive impact on PVAT-derived factors and may restore the balance in their concentration.
Collapse
|
25
|
Motta JM, Rumjanek VM, Mantovani A, Locati M. Tumor-Released Products Promote Bone Marrow-Derived Macrophage Survival and Proliferation. Biomedicines 2021; 9:biomedicines9101387. [PMID: 34680504 PMCID: PMC8533124 DOI: 10.3390/biomedicines9101387] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 09/03/2021] [Accepted: 09/06/2021] [Indexed: 12/23/2022] Open
Abstract
Macrophages play a central role within the tumor microenvironment, with relevant implications for tumor progression. The modulation of their phenotype is one of the mechanisms used by tumors to escape from effective immune responses. This study was designed to analyze the influence of soluble products released by tumors, here represented by the tumor-conditioned media of two tumor cell lines (3LL from Lewis lung carcinoma and MN/MCA from fibrosarcoma), on murine macrophage differentiation and polarization in vitro. Data revealed that tumor-conditioned media stimulated macrophage differentiation but influenced the expression levels of macrophage polarization markers, cytokine production, and microRNAs of relevance for macrophage biology. Interestingly, tumor-derived soluble products supported the survival and proliferation rate of bone marrow precursor cells, an effect observed even with mature macrophages in the presence of M2 but not M1 inducers. Despite presenting low concentrations of macrophage colony-stimulating factor (M-CSF), tumor-conditioned media alone also supported the proliferation of cells to a similar extent as exogenous M-CSF. This effect was only evident in cells positive for the expression of the M-CSF receptor (CD115) and occurred preferentially within the CD16+ subset. Blocking CD115 partially reversed the effect on proliferation. These results suggest that tumors release soluble products that not only promote macrophage development from bone marrow precursors but also stimulate the proliferation of cells with specific phenotypes that could support protumoral functions.
Collapse
Affiliation(s)
- Juliana Maria Motta
- Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (J.M.M.); (V.M.R.)
- Humanitas Clinical and Research Center—IRCCS, 20089 Rozzano, Italy;
| | - Vivian Mary Rumjanek
- Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (J.M.M.); (V.M.R.)
| | | | - Massimo Locati
- Humanitas Clinical and Research Center—IRCCS, 20089 Rozzano, Italy;
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20122 Milan, Italy
- Correspondence:
| |
Collapse
|
26
|
Biagioli M, Fiorucci S. Bile acid activated receptors: Integrating immune and metabolic regulation in non-alcoholic fatty liver disease. LIVER RESEARCH 2021; 5:119-141. [PMID: 39957845 PMCID: PMC11791866 DOI: 10.1016/j.livres.2021.08.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/29/2021] [Accepted: 08/27/2021] [Indexed: 02/06/2023]
Abstract
Bile acids are a family of atypical steroids generated at the interface of liver-intestinal microbiota acting on a ubiquitously expressed family of membrane and nuclear receptors known as bile acid activated receptors. The two best characterized receptors of this family are the nuclear receptor, farnesoid X receptor (FXR) and the G protein-coupled receptor, G protein-coupled bile acid receptor 1 (GPBAR1). FXR and GPBAR1 regulate major aspects of lipid and glucose metabolism, energy balance, autophagy and immunity and have emerged as potential pharmaceutical targets for the treatment of metabolic and inflammatory disorders. Clinical trials in non-alcoholic fatty liver disease (NAFLD), however, have shown that selective FXR agonists cause side effects while their efficacy is partial. Because FXR and GPBAR1 exert additive effects, dual FXR/GPBAR1 ligands have been developed for the treatment of metabolic disorders and are currently advanced to clinical trials. Here, we will review the role of FXR and GPBAR1 agonism in NAFLD and how the two receptors could be exploited to target multiple components of the disease.
Collapse
Affiliation(s)
- Michele Biagioli
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Stefano Fiorucci
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
27
|
Adipose Tissue Immunomodulation and Treg/Th17 Imbalance in the Impaired Glucose Metabolism of Children with Obesity. CHILDREN-BASEL 2021; 8:children8070554. [PMID: 34199040 PMCID: PMC8305706 DOI: 10.3390/children8070554] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/18/2021] [Accepted: 06/25/2021] [Indexed: 12/12/2022]
Abstract
In the last few decades, obesity has increased dramatically in pediatric patients. Obesity is a chronic disease correlated with systemic inflammation, characterized by the presence of CD4 and CD8 T cell infiltration and modified immune response, which contributes to the development of obesity related diseases and metabolic disorders, including impaired glucose metabolism. In particular, Treg and Th17 cells are dynamically balanced under healthy conditions, but imbalance occurs in inflammatory and pathological states, such as obesity. Some studies demonstrated that peripheral Treg and Th17 cells exhibit increased imbalance with worsening of glucose metabolic dysfunction, already in children with obesity. In this review, we considered the role of adipose tissue immunomodulation and the potential role played by Treg/T17 imbalance on the impaired glucose metabolism in pediatric obesity. In the patient care, immune monitoring could play an important role to define preventive strategies of pediatric metabolic disease treatments.
Collapse
|
28
|
Lactoferrin and Immunoglobulin Concentrations in Milk of Gestational Diabetic Mothers. Nutrients 2021; 13:nu13030818. [PMID: 33801292 PMCID: PMC7998843 DOI: 10.3390/nu13030818] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 02/22/2021] [Accepted: 02/25/2021] [Indexed: 01/03/2023] Open
Abstract
Gestational diabetes mellitus (GDM) is associated with an increased risk of having a high-care newborn and has an impact on maternal wellbeing. This study aimed to assess the effect of GDM on the lactoferrin (LF), secretory immunoglobulin A (SIgA), immunoglobulin G (IgG), and immunoglobulin M (IgM) concentrations in early colostrum, colostrum, and transitional milk samples of hyperglycemic (n = 53) and normoglycemic (n = 49) mothers using enzyme-linked immunosorbent assay (ELISA). The concentrations of milk lactoferrin and SIgA, but not IgG and IgM, from hyperglycemic and normoglycemic mothers, showed a similar negative correlation with lactation from the first to the fifteenth day. Apart from early colostral IgG, there were no differences in concentrations of LF and immunoglobulins in milk from hyperglycemic and normoglycemic mothers. For hyperglycemia compensated by diet (GDM G1) or insulin treatment (GDM G2), slight differences were seen for LF and IgG, but not for SIgA and IgM, during an early stage of lactation only. Early colostral IgG and colostral LF of insulin-treated mothers were higher (10.01 ± 4.48 mg/L and 11.50 ± 0.58 g/L, respectively) than for diet-control diabetic mothers (7.65 ± 5.67 mg/L and 8.05 ± 1.38 g/L, respectively). GDM of mothers does not have a significant impact on immunological quality of early milk.
Collapse
|
29
|
Sinha SK, Shaheen M, Singh VR, Rajavashisth TB, Pan D, Sun L, Norris KC, Nicholas SB. How Clinically Relevant Is C-Reactive Protein for Blacks with Metabolic Syndrome to Predict Microalbuminuria? Metab Syndr Relat Disord 2021; 19:39-47. [PMID: 32896227 PMCID: PMC7891189 DOI: 10.1089/met.2019.0121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Background: The metabolic syndrome (MetS) is associated with elevated urinary albumin (UA) excretion and C-reactive protein (CRP). However, potential differences in CRP levels on the association between individual components of the MetS and microalbuminuria (MA; 30-300 μg/mL) and/or UA (0-300 μg/mL) by race/ethnicity is unknown. Methods: We analyzed National Health and Nutrition Examination Surveys (NHANES) data, (1999-2010) for adults (≥20 years of age) with the MetS (N = 5700). The Sobel-Goodman mediation test examined the influence of CRP on the association between individual MetS components and both MA and UA by race/ethnicity. We applied machine learning models to predict UA. Results: CRP mediated the association between waist circumference (WC) and MA in Whites and Hispanics but not in Blacks. However, in general, the proportion of the total effect of MetS components on UA, mediated by CRP, was: 11% for high-density lipoprotein cholesterol (HDL-C) and 40% for WC (P < 0.001). In contrast to MA, the mediation effect of CRP for WC and UA was highest for Blacks (94%) compared with Whites (55%) or Hispanics (18%), P < 0.05. The prediction of an elevated UA concentration was increased in Blacks (∼51%) with the MetS when CRP was added to the random forest model. Conclusions: CRP mediates the association between UA and both HDL-C and WC in Whites and Blacks and between UA and WC in Hispanics. Moreover, the machine learning approach suggests that the incorporation of CRP may improve model prediction of UA in Blacks. These findings may favor screening for CRP in persons with the MetS, particularly in Blacks.
Collapse
Affiliation(s)
- Satyesh K. Sinha
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
- Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California, USA
| | - Magda Shaheen
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
- Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California, USA
| | | | - Tripathi B. Rajavashisth
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
- Molecular Biology Unit, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Deyu Pan
- Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California, USA
| | - Ling Sun
- Division of Nephrology, Xuzhou Central Hospital, Medical College of Southeast University, Xuzhou, China
| | - Keith C. Norris
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Susanne B. Nicholas
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| |
Collapse
|
30
|
Li X, Frazier JA, Spahiu E, McPherson M, Miller RA. Muscle-dependent regulation of adipose tissue function in long-lived growth hormone-mutant mice. Aging (Albany NY) 2020; 12:8766-8789. [PMID: 32464603 PMCID: PMC7288969 DOI: 10.18632/aging.103380] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 05/14/2020] [Indexed: 01/24/2023]
Abstract
Altered adipose tissue may contribute to the longevity of Snell dwarf and growth hormone receptor (GHR) knock-out mice. We report here that white (WAT) and brown (BAT) fat have elevated UCP1 in both kinds of mice, and that adipocytes in WAT depots turn beige/brown. These imply increased thermogenesis and are expected to lead to improved glucose control. Both kinds of long-lived mice show lower levels of inflammatory M1 macrophages and higher levels of anti-inflammatory M2 macrophages in BAT and WAT, with correspondingly lower levels of TNFα, IL-6, and MCP1. Experiments with mice with tissue-specific disruption of GHR showed that these adipocyte and macrophage changes were not due to hepatic IGF1 production nor to direct GH effects on adipocytes, but instead reflect GH effects on muscle. Muscles deprived of GH signals, either globally (GKO) or in muscle only (MKO), produce higher levels of circulating irisin and its precursor FNDC5. The data thus suggest that the changes in adipose tissue differentiation and inflammatory status seen in long-lived mutant mice reflect interruption of GH-dependent irisin inhibition, with consequential effects on metabolism and thermogenesis.
Collapse
Affiliation(s)
- Xinna Li
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan 48109, USA
| | - Jacquelyn A. Frazier
- College of Literature, Sciences, and The Arts, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Edward Spahiu
- College of Literature, Sciences, and The Arts, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Madaline McPherson
- College of Literature, Sciences, and The Arts, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Richard A. Miller
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan 48109, USA,University of Michigan Geriatrics Center, Ann Arbor, Michigan 48109, USA
| |
Collapse
|
31
|
Use of Chlorogenic Acid against Diabetes Mellitus and Its Complications. J Immunol Res 2020; 2020:9680508. [PMID: 32566690 PMCID: PMC7275206 DOI: 10.1155/2020/9680508] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/27/2020] [Accepted: 05/04/2020] [Indexed: 01/11/2023] Open
Abstract
Chlorogenic acid (CA) is a phenolic compound commonly found in human plant-based diets. CA is the main component of many traditional Chinese medicine preparations, and in recent years, it has been found to have hypoglycemic, hypolipidemic, anti-inflammatory, antioxidant, and other pharmacological properties. Specifically, CA relieves the effects of, and prevents, diabetes mellitus (DM). In addition, CA is also beneficial against complications arising from DM, such as diabetic nephropathy (DN), diabetic retinopathy (DR), and diabetic peripheral neuropathy (DPN). Herein, we review the use of CA in the prevention and treatment of DM and its complications, providing a background for further research and medical uses.
Collapse
|
32
|
Orliaguet L, Dalmas E, Drareni K, Venteclef N, Alzaid F. Mechanisms of Macrophage Polarization in Insulin Signaling and Sensitivity. Front Endocrinol (Lausanne) 2020; 11:62. [PMID: 32140136 PMCID: PMC7042402 DOI: 10.3389/fendo.2020.00062] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 01/30/2020] [Indexed: 12/11/2022] Open
Abstract
Type-2 diabetes (T2D) is a disease of two etiologies: metabolic and inflammatory. At the cross-section of these etiologies lays the phenomenon of metabolic inflammation. Whilst metabolic inflammation is characterized as systemic, a common starting point is the tissue-resident macrophage, who's successful physiological or aberrant pathological adaptation to its microenvironment determines disease course and severity. This review will highlight the key mechanisms in macrophage polarization, inflammatory and non-inflammatory signaling that dictates the development and progression of insulin resistance and T2D. We first describe the known homeostatic functions of tissue macrophages in insulin secreting and major insulin sensitive tissues. Importantly we highlight the known mechanisms of aberrant macrophage activation in these tissues and the ways in which this leads to impairment of insulin sensitivity/secretion and the development of T2D. We next describe the cellular mechanisms that are known to dictate macrophage polarization. We review recent progress in macrophage bio-energetics, an emerging field of research that places cellular metabolism at the center of immune-effector function. Importantly, following the advent of the metabolically-activated macrophage, we cover the known transcriptional and epigenetic factors that canonically and non-canonically dictate macrophage differentiation and inflammatory polarization. In closing perspectives, we discuss emerging research themes and highlight novel non-inflammatory or non-immune roles that tissue macrophages have in maintaining microenvironmental and systemic homeostasis.
Collapse
Affiliation(s)
- Lucie Orliaguet
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, Paris, France
| | - Elise Dalmas
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, Paris, France
| | - Karima Drareni
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, Paris, France
- Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, United States
| | - Nicolas Venteclef
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, Paris, France
| | - Fawaz Alzaid
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, Paris, France
| |
Collapse
|
33
|
Jiang W, Ma T, Zhang C, Tang X, Xu Q, Meng X, Ma T. Identification of urinary candidate biomarkers of cisplatin-induced nephrotoxicity in patients with carcinoma. J Proteomics 2020; 210:103533. [DOI: 10.1016/j.jprot.2019.103533] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 08/23/2019] [Accepted: 09/19/2019] [Indexed: 10/25/2022]
|
34
|
Gibbs JL, Dallon BW, Lewis JB, Walton CM, Arroyo JA, Reynolds PR, Bikman BT. Diesel Exhaust Particle Exposure Compromises Alveolar Macrophage Mitochondrial Bioenergetics. Int J Mol Sci 2019; 20:ijms20225598. [PMID: 31717476 PMCID: PMC6888061 DOI: 10.3390/ijms20225598] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/29/2019] [Accepted: 11/06/2019] [Indexed: 12/15/2022] Open
Abstract
Diesel exhaust particles (DEPs) are known pathogenic pollutants that constitute a significant quantity of air pollution. Given the ubiquitous presence of macrophages throughout the body, including the lungs, as well as their critical role in tissue and organismal metabolic function, we sought to determine the effect of DEP exposure on macrophage mitochondrial function. Following daily DEP exposure in mice, pulmonary macrophages were isolated for mitochondrial analyses, revealing reduced respiration rates and dramatically elevated H2O2 levels. Serum ceramides and inflammatory cytokines were increased. To determine the degree to which the changes in mitochondrial function in macrophages were not dependent on any cross-cell communication, primary pulmonary murine macrophages were used to replicate the DEP exposure in a cell culture model. We observed similar changes as seen in pulmonary macrophages, namely diminished mitochondrial respiration, but increased H2O2 production. Interestingly, when treated with myriocin to inhibit ceramide biosynthesis, these DEP-induced mitochondrial changes were mitigated. Altogether, these data suggest that DEP exposure may compromise macrophage mitochondrial and whole-body function via pathologic alterations in macrophage ceramide metabolism.
Collapse
Affiliation(s)
- Jonathan L. Gibbs
- Metabolism Research Lab, Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT 84602, USA
| | - Blake W. Dallon
- Metabolism Research Lab, Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT 84602, USA
| | - Joshua B. Lewis
- Lung and Placental Research Lab, Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT 84602, USA
| | - Chase M. Walton
- Metabolism Research Lab, Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT 84602, USA
| | - Juan A. Arroyo
- Lung and Placental Research Lab, Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT 84602, USA
| | - Paul R. Reynolds
- Lung and Placental Research Lab, Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT 84602, USA
| | - Benjamin T. Bikman
- Metabolism Research Lab, Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT 84602, USA
- Correspondence:
| |
Collapse
|
35
|
Role and mechanism of the Th17/Treg cell balance in the development and progression of insulin resistance. Mol Cell Biochem 2019; 459:183-188. [PMID: 31218568 PMCID: PMC6679830 DOI: 10.1007/s11010-019-03561-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 05/27/2019] [Indexed: 12/23/2022]
Abstract
The pathogenic mechanism of insulin resistance and associated diseases such as metabolic syndrome and diabetes remains unclear. Since inflammatory cytokines secreted by T cells play an important role in immune system homeostasis, we evaluated the role of interleukin-6 (IL-6) and the Th17/Treg balance in insulin sensitivity and the underlying mechanism in a rat model. After establishing an insulin-resistant rat model, the rats were injected with anti-mouse IL-6R receptor antibody (MR16-1) to block IL-6. Adipose tissue and blood samples were obtained for the analysis of cytokines, Th17 and Treg markers, and insulin sensitivity blood parameters, for comparisons with those of the normal control group, IL-6-blocked control group, and insulin resistance control group. In the insulin resistance control group, the expression levels of IL-6, RORγt, and IL-17 increased, whereas those of IL-10, FoxP3, and CD4+CD25+Treg decreased. Insulin sensitivity decreased, whereas glucose, total serum cholesterol, triglycerides, and free fatty acid levels significantly increased. However, the completely opposite effects for all parameters were detected in the insulin resistance IL-6-blocked group. Insulin resistance can cause inflammation and an imbalance in Th17 cells/Treg cells. IL-6 can restore this imbalance and play an important role in the development and progression of insulin resistance.
Collapse
|
36
|
Drareni K, Gautier JF, Venteclef N, Alzaid F. Transcriptional control of macrophage polarisation in type 2 diabetes. Semin Immunopathol 2019; 41:515-529. [PMID: 31049647 DOI: 10.1007/s00281-019-00748-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 04/18/2019] [Indexed: 01/09/2023]
Abstract
Type-2 diabetes (T2D) is considered today as an inflammatory disease. Inflammatory processes in T2D are orchestrated by macrophage activation in different organs. Macrophages undergo classical M1 pro-inflammatory or alternative M2 anti-inflammatory activation in response to tissue microenvironmental signals. These subsets of macrophages are characterised by their expression of cell surface markers, secreted cytokines and chemokines. Transcriptional regulation is central to the polarisation of macrophages, and several major pathways have been described as essential to promote the expression of specific genes, which dictate the functional polarisation of macrophages. In this review, we summarise the current knowledge of transcriptional control of macrophage polarisation and the role this plays in development of insulin resistance.
Collapse
Affiliation(s)
- Karima Drareni
- Cordeliers Research Centre, INSERM, Immunity and Metabolism in Diabetes Laboratory, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, F-75006, Paris, France
| | - Jean-François Gautier
- Cordeliers Research Centre, INSERM, Immunity and Metabolism in Diabetes Laboratory, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, F-75006, Paris, France.,Lariboisière Hospital, AP-HP, Diabetology Department, University of Paris Diderot, Paris, France
| | - Nicolas Venteclef
- Cordeliers Research Centre, INSERM, Immunity and Metabolism in Diabetes Laboratory, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, F-75006, Paris, France.
| | - Fawaz Alzaid
- Cordeliers Research Centre, INSERM, Immunity and Metabolism in Diabetes Laboratory, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, F-75006, Paris, France.
| |
Collapse
|
37
|
The role of macrophages in nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. Nat Rev Gastroenterol Hepatol 2019; 16:145-159. [PMID: 30482910 DOI: 10.1038/s41575-018-0082-x] [Citation(s) in RCA: 632] [Impact Index Per Article: 105.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) and its inflammatory and often progressive subtype nonalcoholic steatohepatitis (NASH) are becoming the leading cause of liver-related morbidity and mortality worldwide, and a primary indication for liver transplantation. The pathophysiology of NASH is multifactorial and not yet completely understood; however, innate immunity is a major contributing factor in which liver-resident macrophages (Kupffer cells) and recruited macrophages play a central part in disease progression. In this Review, we assess the evidence for macrophage involvement in the development of steatosis, inflammation and fibrosis in NASH. In this process, not only the polarization of liver macrophages towards a pro-inflammatory phenotype is important, but adipose tissue macrophages, especially in the visceral compartment, also contribute to disease severity and insulin resistance. Macrophage activation is mediated by factors such as endotoxins and translocated bacteria owing to increased intestinal permeability, factors released from damaged or lipoapoptotic hepatocytes, as well as alterations in gut microbiota and defined nutritional components, including certain free fatty acids, cholesterol and their metabolites. Reflecting the important role of macrophages in NASH, we also review studies investigating drugs that target macrophage recruitment to the liver, macrophage polarization and their inflammatory effects as potential treatment options for patients with NASH.
Collapse
|
38
|
Melekoglu R, Ciftci O, Celik E, Yilmaz E, Bastemur AG. Evaluation of second trimester amniotic fluid ADAMTS4, ADAMTS5, interleukin-6 and tumor necrosis factor-α levels in patients with gestational diabetes mellitus. J Obstet Gynaecol Res 2019; 45:824-829. [PMID: 30623540 DOI: 10.1111/jog.13914] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 12/08/2018] [Indexed: 01/04/2023]
Abstract
AIM To test the hypothesis that altered A Disintegrin and Metalloproteinase Domains with Thrombospondins motifs (ADAMTS) is implicated in the etiopathogenesis of gestational diabetes mellitus (GDM). METHODS All pregnant women who underwent elective amniocentesis for karyotype analysis between January 1, 2016, and January 1, 2018, were included in this study. From this cohort, the study group consisting of 20 patients diagnosed with GDM was selected and compared against a control group consisting of 20 age- and body mass index (BMI)-matched patients without GDM. ADAMTS4, ADAMTS5, interleukin-6 (IL-6) and tumor necrosis factor alpha (TNF-α) levels were compared in the second trimester amniotic fluid of patients with GDM and normoglycemic pregnant women. RESULTS No significant differences were observed between GDM and control groups regarding age, BMI, gestational age at amniocentesis and indication for amniocentesis. Mean amniotic fluid ADAMTS4 and ADAMTS5 levels were significantly increased in the GDM group compared with the control group (253.5 ± 18.7 pg/mL and 188.5 ± 21.3 pg/mL, P < 0.001; 192.9 ± 16.4 pg/mL and 154.8 ± 19.9 pg/mL, P = 0.021, respectively). Significant increases in IL-6 and TNF-α levels were also detected in the amniotic fluid of GDM patients relative to controls (136.2 ± 17.3 pg/mL and 98.3 ± 11.5 pg/mL, P < 0.001; 154.2 ± 12.5 pg/mL and 86.2 ± 10.8 pg/mL, P < 0.001, respectively). CONCLUSION The data presented here suggest that increased levels of ADAMTS4, ADAMTS5, IL-6 and TNF-α may play an important role in the progression of GDM.
Collapse
Affiliation(s)
- Rauf Melekoglu
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Inonu, Malatya, Turkey
| | - Osman Ciftci
- Department of Medical Pharmacology, Faculty of Medicine, University of Pamukkale, Denizli, Turkey
| | - Ebru Celik
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Koc, İstanbul, Turkey
| | - Ercan Yilmaz
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Inonu, Malatya, Turkey
| | - Ayse G Bastemur
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Inonu, Malatya, Turkey
| |
Collapse
|
39
|
ELSHAHIDI MH. Outdoor Air Pollution and Gestational Diabetes Mellitus: A Systematic Review and Meta-Analysis. IRANIAN JOURNAL OF PUBLIC HEALTH 2019; 48:9-19. [PMID: 30847307 PMCID: PMC6401584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
BACKGROUND During the past 20 years, the prevalence of gestational diabetes mellitus (GDM) has increased by ∼10%-100% in several race/ethnicity groups. There is an association between ambient air pollution (AAP) and GDM. This study aimed to summarize the evidence about the association between AAP and GDM. METHODS PubMed, Embase, Scopus, Web of Science and Cochrane Library were searched from inception till Oct 2017. Studies about the association between ambient air pollutants levels and GDM were included. Pooled effect estimates and their 95% confidence interval (CI) were calculated using R. RESULTS Eight studies met the inclusion criteria. The odds of developing GDM upon exposure to CO (per 1 ppm), NO (per 1 ppb), NO2 (per 10 μg/m3), NOx (per 1 ppb), O3 (per 10 ppb), SO2 (per 10 ppb), PM10 (per 10 μg/m3) and PM2.5 (per 10 μg/m3) were 1.47 (95% CI 0.88-2.06), 1.04 (95% CI 1.03-1.06), 1 (95% CI 0.93-1.08), 1.02 (95% CI 1-1.04), 1.05 (95% CI 0.94-1.16), 1.39 (95% CI 1.04-1.73), 0.97 (95% CI 0.94-0.99) and 1.12 (95% CI 0.93-1.31), respectively. CONCLUSION The current literature showed evidence for an association between AAP and GDM. However, further well-designed studies are needed.
Collapse
|
40
|
Positive Correlation of Serum N-Acetyl- β-hexosaminidase with Markers of Atherosclerosis in Diabetes Type 2 Patients with Mild Symptoms of Depression and Anxiety. DISEASE MARKERS 2018; 2018:1760592. [PMID: 30026880 PMCID: PMC6031159 DOI: 10.1155/2018/1760592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Accepted: 05/25/2018] [Indexed: 11/22/2022]
Abstract
Background Analysis of the correlation between diabetes type 2 (DT2) and serum N-acetyl-β-hexosaminidase (HEX) activity with parameters of fat metabolism and symptoms of anxiety and depression. Material and Method The study was performed using a random sample of 40 DT2 patients (22 women and 18 men) between the ages of 43 and 71 (median 59) and 40 control persons (28 women and 12 men) between the ages of 18 and 64 (median 46). The activity of HEX was determined by a colorimetric method. The activity of the serum exoglycosidase was expressed in pkat/mL. Each participant underwent Hamilton tests, to evaluate level of anxiety and depression. Additionally, the HEX activity and concentration of particular lipidograms were monitored using a blood sample from each participant. Results In DT2 patients, a significant positive correlation was found between serum HEX activity and the concentration of serum cholesterol LDL fractions, triacylglycerols (TAG), and Castelligro atherogenic indexes. A significantly increased level of anxiety and depression in comparison to the control group was found as well. Conclusion Serum HEX activity in DT2 patients is a better marker of atherosclerosis than serum total cholesterol level in persons with mild symptoms of depression and anxiety. In DT2 patients, a routine testing of anxiety and depression is recommended. Early detection of these disorders creates the possibility for treatment, an improvement in a patient's quality of life, and the overall longevity of DT2 patients.
Collapse
|
41
|
Gonzalez LL, Garrie K, Turner MD. Type 2 diabetes - An autoinflammatory disease driven by metabolic stress. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3805-3823. [PMID: 30251697 DOI: 10.1016/j.bbadis.2018.08.034] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 08/27/2018] [Indexed: 02/06/2023]
Abstract
Type 2 diabetes has traditionally been viewed as a metabolic disorder characterised by chronic high glucose levels, insulin resistance, and declining insulin secretion from the pancreas. Modern lifestyle, with abundant nutrient supply and reduced physical activity, has resulted in dramatic increases in the rates of obesity-associated disease conditions, including diabetes. The associated excess of nutrients induces a state of systemic low-grade chronic inflammation that results from production and secretion of inflammatory mediators from the expanded pool of activated adipocytes. Here, we review the mechanisms by which obesity induces adipose tissue dysregulation, detailing the roles of adipose tissue secreted factors and their action upon other cells and tissues central to glucose homeostasis and type 2 diabetes. Furthermore, given the emerging importance of adipokines, cytokines and chemokines in disease progression, we suggest that type 2 diabetes should now be viewed as an autoinflammatory disease, albeit one that is driven by metabolic dysregulation.
Collapse
Affiliation(s)
- Laura L Gonzalez
- Interdisciplinary Biomedical Research Centre, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham NG11 8NS, United Kingdom
| | - Karin Garrie
- Interdisciplinary Biomedical Research Centre, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham NG11 8NS, United Kingdom
| | - Mark D Turner
- Interdisciplinary Biomedical Research Centre, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham NG11 8NS, United Kingdom.
| |
Collapse
|
42
|
Reich D, Gallucci G, Tong M, de la Monte SM. Therapeutic Advantages of Dual Targeting of PPAR-δ and PPAR-γ in an Experimental Model of Sporadic Alzheimer's Disease. ACTA ACUST UNITED AC 2018; 5. [PMID: 30705969 PMCID: PMC6350901 DOI: 10.13188/2376-922x.1000025] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Background: Alzheimer’s disease (AD) is associated with progressive impairments in brain responsiveness to insulin and insulin-like growth factor (IGF). Although deficiencies in brain insulin and IGF could be ameliorated with trophic factors such as insulin, impairments in receptor expression, binding, and tyrosine kinase activation require alternative strategies. Peroxisome proliferator-activated receptor (PPAR) agonists target genes downstream of insulin/IGF stimulation. Furthermore, their anti-oxidant and anti-inflammatory effects address other pathologies contributing to neurodegeneration. Objectives: The goal of this research was to examine effects of dual delivery of L165, 041 (PPAR-δ) and F-L-Leu (PPAR-γ) agonists for remediating in the early stages of neurodegeneration. Model: Experiments were conducted using frontal lobe slice cultures from an intracerebral Streptozotocin (i.c. STZ) rat model of AD. Results: PPAR-δ+ PPAR-γ agonist treatments increased indices of neuronal and myelin maturation, and mitochondrial proliferation and function, and decreased neuroinflammation, AβPP-Aβ, neurotoxicity, ubiquitin, and nitrosative stress, but failed to restore choline acetyl transferase expression and adversely increased HNE(lipid peroxidation) and acetylcholinesterase, which would have further increased stress and reduced cholinergic function in the STZ brain cultures. Conclusion: PPAR-δ + PPAR-γ agonist treatments have substantial positive early therapeutic targeting effects on AD-associated molecular and biochemical brain pathologies. However, additional or alternative strategies may be needed to optimize disease remediation during the initial phases of treatment.
Collapse
Affiliation(s)
- D Reich
- Brandeis University, Waltham University, USA
| | - G Gallucci
- Department of Medicine, University of Brown University, USA
| | - M Tong
- Department of Medicine, University of Brown University, USA
| | - S M de la Monte
- Department of Medicine, University of Brown University, USA.,Departments of Neurology, University of Brown University, USA
| |
Collapse
|
43
|
Shavva VS, Bogomolova AM, Efremov AM, Trofimov AN, Nikitin AA, Babina AV, Nekrasova EV, Dizhe EB, Oleinikova GN, Missyul BV, Orlov SV. Insulin downregulates C3 gene expression in human HepG2 cells through activation of PPARγ. Eur J Cell Biol 2018; 97:204-215. [DOI: 10.1016/j.ejcb.2018.03.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 03/07/2018] [Accepted: 03/08/2018] [Indexed: 01/31/2023] Open
|
44
|
Hakansson AP, Orihuela CJ, Bogaert D. Bacterial-Host Interactions: Physiology and Pathophysiology of Respiratory Infection. Physiol Rev 2018; 98:781-811. [PMID: 29488821 PMCID: PMC5966719 DOI: 10.1152/physrev.00040.2016] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 09/08/2017] [Accepted: 09/11/2017] [Indexed: 02/06/2023] Open
Abstract
It has long been thought that respiratory infections are the direct result of acquisition of pathogenic viruses or bacteria, followed by their overgrowth, dissemination, and in some instances tissue invasion. In the last decades, it has become apparent that in contrast to this classical view, the majority of microorganisms associated with respiratory infections and inflammation are actually common members of the respiratory ecosystem and only in rare circumstances do they cause disease. This suggests that a complex interplay between host, environment, and properties of colonizing microorganisms together determines disease development and its severity. To understand the pathophysiological processes that underlie respiratory infectious diseases, it is therefore necessary to understand the host-bacterial interactions occurring at mucosal surfaces, along with the microbes inhabiting them, during symbiosis. Current knowledge regarding host-bacterial interactions during asymptomatic colonization will be discussed, including a plausible role for the human microbiome in maintaining a healthy state. With this as a starting point, we will discuss possible disruptive factors contributing to dysbiosis, which is likely to be a key trigger for pathobionts in the development and pathophysiology of respiratory diseases. Finally, from this renewed perspective, we will reflect on current and potential new approaches for treatment in the future.
Collapse
Affiliation(s)
- A P Hakansson
- Division of Experimental Infection Medicine, Department of Translational Medicine, Lund University , Lund , Sweden ; Department of Microbiology, University of Alabama at Birmingham , Birmingham, Alabama ; and Center for Inflammation Research, Queens Medical Research Institute, University of Edinburgh , Edinburgh , United Kingdom
| | - C J Orihuela
- Division of Experimental Infection Medicine, Department of Translational Medicine, Lund University , Lund , Sweden ; Department of Microbiology, University of Alabama at Birmingham , Birmingham, Alabama ; and Center for Inflammation Research, Queens Medical Research Institute, University of Edinburgh , Edinburgh , United Kingdom
| | - D Bogaert
- Division of Experimental Infection Medicine, Department of Translational Medicine, Lund University , Lund , Sweden ; Department of Microbiology, University of Alabama at Birmingham , Birmingham, Alabama ; and Center for Inflammation Research, Queens Medical Research Institute, University of Edinburgh , Edinburgh , United Kingdom
| |
Collapse
|
45
|
de la Monte SM, Tong M, Schiano I, Didsbury J. Improved Brain Insulin/IGF Signaling and Reduced Neuroinflammation with T3D-959 in an Experimental Model of Sporadic Alzheimer's Disease. J Alzheimers Dis 2018; 55:849-864. [PMID: 27802237 DOI: 10.3233/jad-160656] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is associated with progressive impairments in brain insulin, insulin-like growth factor (IGF), and insulin receptor substrate (IRS) signaling through Akt pathways that regulate neuronal growth, survival, metabolism, and plasticity. The intracerebral streptozotocin (i.c. STZ) model replicates the full range of abnormalities in sporadic AD. T3D-959, an orally active PPAR-delta/gamma agonist remediates neurocognitive deficits and AD neuropathology in the i.c. STZ model. OBJECTIVE This study characterizes the effects of T3D-959 on AD biomarkers, insulin/IGF/IRS signaling through Akt pathways, and neuroinflammation in an i.c. STZ model. METHODS Long Evans rats were treated with i.c. STZ or saline, followed by daily oral doses of T3D-959 (1 mg/kg) or saline initiated 1 day (T3D-959-E) or 7 days (T3D-959-L) later through Experimental Day 28. Protein and phospho-protein expression and pro-inflammatory cytokine activation were measured in temporal lobe homogenates by duplex or multiplex bead-based ELISAs. RESULTS i.c. STZ treatments caused neurodegeneration with increased pTau, AβPP, Aβ42, ubiquitin, and SNAP-25, and reduced levels of synaptophysin, IGF-1 receptor (R), IRS-1, Akt, p70S6K, mTOR, and S9-GSK-3β. i.c. STZ also broadly increased neuroinflammation. T3D-959 abrogated or reduced most of the AD neuropathological and biomarker abnormalities, increased/normalized IGF-1R, IRS-1, Akt, p70S6K, and S9-GSK-3β, and decreased expression of multiple pro-inflammatory cytokines. T3D-959-E or -L effectively restored insulin/IGF signaling, whereas T3D-959-L more broadly resolved neuroinflammation. CONCLUSION AD remediating effects of T3D-959 are potentially due to enhanced expression of key insulin/IGF signaling proteins and inhibition of GSK-3β and neuroinflammation. These effects lead to reduced neurodegeneration, cognitive impairment, and AD biomarker levels in the brain.
Collapse
Affiliation(s)
- Suzanne M de la Monte
- Liver Research Center, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA.,Division of Gastroenterology, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA.,Division of Neuropathology, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA.,Department of Medicine, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA.,Department of Pathology, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA.,Department of Neurology, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA.,Department of Neurosurgery, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Ming Tong
- Liver Research Center, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA.,Division of Gastroenterology, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA.,Department of Medicine, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Irio Schiano
- Quinnipiac University, Biomedical Sciences Department in Hamden, CT, USA
| | - John Didsbury
- T3D Therapeutics, Inc., Research Triangle Park, NC, USA
| |
Collapse
|
46
|
The In Vitro and In Vivo Anti-Inflammatory Effects of a Phthalimide PPAR-γ Agonist. Mar Drugs 2017; 15:md15010007. [PMID: 28054961 PMCID: PMC5295227 DOI: 10.3390/md15010007] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 12/05/2016] [Accepted: 12/09/2016] [Indexed: 01/09/2023] Open
Abstract
Previously, the authors found that 4-hydroxy-2-(4-hydroxyphenethyl) isoindoline-1,3-dione (PD1) (a phthalimide analogue) bound to and activated peroxisome proliferator-activated receptor-γ (PPAR-γ). Since PPAR-γ suppresses inflammatory responses, the present study was undertaken to investigate the anti-inflammatory effects of PD1. In lipopolysaccharide (LPS)-stimulated murine RAW264.7 macrophages, PD1 suppressed the inductions of pro-inflammatory factors, including inducible nitric oxide synthase (iNOS), nitric oxide (NO), cyclooxygenase 2 (COX-2), tumor necrosis factor α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6). Concomitantly, PD1 enhanced the expressions of anti-inflammatory factors, such as arginase-1 and interleukin-10 (IL-10), and suppressed LPS-evoked nuclear factor kappa B (NF-κB) p65 subunit phosphorylation in macrophages. In addition, PPAR-γ activated by PD1 was intensively translocated to the nucleus. These observations suggest that the anti-inflammatory mechanism of PD1 involves inhibition of the NF-κB pathway. In a subsequent in vivo animal experiment conducted using a carrageenan-induced acute inflammatory rat paw edema model, intraperitoneal injection of PD1 significantly reduced paw swelling. Histological analysis of rat paw tissue sections revealed less infiltration of immune cells in PD1-pretreated animals. These findings suggest that PD1 be viewed as a lead compound for the development of novel anti-inflammatory therapeutics.
Collapse
|
47
|
Mohammadi A, Fallah H, Gholamhosseinian A. Antihyperglycemic Effect of Rosa Damascena is Mediated by PPAR.γ Gene Expression in Animal Model of Insulin Resistance. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2017; 16:1080-1088. [PMID: 29201096 PMCID: PMC5610762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Insulin resistance is a condition in which insulin signaling and action are impaired in insulin sensitive tissues and result in hyperglycemia, hyperlipidemia, and type 2 diabetes mellitus. Our previous studies have shown that Rosa damascena has antihyperglycemic effects on diabetic and normal rats. Therefore, we conducted a study to evaluate the effect of this medicinal plant on insulin sensitivity in rats. This study was performed on high fructose diet insulin resistant rats and pioglitazone, an insulin sensitizing drug, was used as a positive control. Insulin resistance was developed in animals by high fructose diet within six weeks. Then, Rosa damascena extract and pioglitazone were administered by gavage for two weeks and results were compared with two control groups. After treatment period, serum glucose, insulin, adiponectin, triglyceride, and cholesterol were assayed in fasting state. Plasma free fatty acid profile was analyzed by GC. Liver PPAR.γ and muscle GLUT.4 gene expressions were assessed by real time PCR and western blotting. Animals were treated with rosa damascena extract showed levels of insulin (42 ± 2.7 pmol/L). adiponectin (5.6±0.17 μg/mL). glucose (129±4.7 mg/dL). and triglyceride (75 ± 9 mg/dl) which were significantly improved as compared with control group insulin (137 ± 34 pmol/L), adiponectin (3.9±0.15 μg/mL). glucose (187±15 mg/dL). and triglycerides (217±18 mg/dL). PPARγ protein level was also significantly increased in Rosa damascene treated group. Our results demonstrated that rosa damascena extract has useful effects on insulin resistant animals and by increasing insulin sensitivity can be considered as a potential agent in control of diabetes.
Collapse
|
48
|
de la Monte SM. Insulin Resistance and Neurodegeneration: Progress Towards the Development of New Therapeutics for Alzheimer's Disease. Drugs 2017; 77:47-65. [PMID: 27988872 PMCID: PMC5575843 DOI: 10.1007/s40265-016-0674-0] [Citation(s) in RCA: 203] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) should be regarded as a degenerative metabolic disease caused by brain insulin resistance and deficiency, and overlapping with the molecular, biochemical, pathophysiological, and metabolic dysfunctions in diabetes mellitus, non-alcoholic fatty liver disease, and metabolic syndrome. Although most of the diagnostic and therapeutic approaches over the past several decades have focused on amyloid-beta (Aβ42) and aberrantly phosphorylated tau, which could be caused by consequences of brain insulin resistance, the broader array of pathologies including white matter atrophy with loss of myelinated fibrils and leukoaraiosis, non-Aβ42 microvascular disease, dysregulated lipid metabolism, mitochondrial dysfunction, astrocytic gliosis, neuro-inflammation, and loss of synapses vis-à-vis growth of dystrophic neurites, is not readily accounted for by Aβ42 accumulations, but could be explained by dysregulated insulin/IGF-1 signaling with attendant impairments in signal transduction and gene expression. This review covers the diverse range of brain abnormalities in AD and discusses how insulins, incretins, and insulin sensitizers could be utilized to treat at different stages of neurodegeneration.
Collapse
Affiliation(s)
- Suzanne M de la Monte
- Department of Neurology, Rhode Island Hospital, and the Alpert Medical School of Brown University, Pierre Galletti Research Building, 55 Claverick Street, Room 419, Providence, RI, 02903, USA.
- Department of Neurosurgery, Rhode Island Hospital, and the Alpert Medical School of Brown University, Providence, RI, USA.
- Department of Neuropathology, Rhode Island Hospital, and the Alpert Medical School of Brown University, Providence, RI, USA.
- Department of Pathology, Rhode Island Hospital, and the Alpert Medical School of Brown University, Providence, RI, USA.
| |
Collapse
|
49
|
Tong M, Deochand C, Didsbury J, de la Monte SM. T3D-959: A Multi-Faceted Disease Remedial Drug Candidate for the Treatment of Alzheimer's Disease. J Alzheimers Dis 2016; 51:123-38. [PMID: 26836193 DOI: 10.3233/jad-151013] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND T3D-959, a dual PPAR-δ/PPAR γ nuclear receptor agonist and former diabetes drug candidate, has been repositioned as an Alzheimer's disease (AD)-modifying therapy. OBJECTIVE This study examines the effectiveness and mechanisms of T3D-959's therapeutic effects using in vivo and ex vivo rat models of sporadic AD. METHODS A sporadic AD model was generated by intracerebral (i.c.) administration of streptozotocin (STZ). Control and i.c. STZ treated rats were gavaged with saline or T3D-959 (0.3 to 3.0 mg/kg/day) for 28 days. Spatial learning and memory were evaluated using the Morris water maze test. Frontal lobe slice cultures generated 24 hours after i.c. STZ or vehicle were used to study early effects of T3D-959 (0.5-1.0 μM) on viability and molecular markers of AD. RESULTS T3D-959 significantly improved spatial learning and memory in i.c STZ-treated rats. Mechanistically, T3D-959 significantly improved culture viability and brain morphology, reduced levels of oxidative stress and Aβ, and normalized expression of phospho-tau, choline acetyltransferase, and myelin-associated glycoprotein. Protective effects occurred even at the lowest tested dose of T3D-959. CONCLUSIONS Pre-clinical proof of concept has been demonstrated that T3D-959 can improve multiple pathologies of AD resulting in significant improvements in cognitive function and molecular and biochemical indices of neurodegeneration. These results support the theses that (1) effective disease modification in AD can be achieved by targeting relevant nuclear receptors, and (2) treating AD as a metabolic disease has the potential to be disease remedial. A Phase 2a trial of T3D-959 in mild-to-moderate AD patients has been initiated (ClinicalTrials.gov identifier NCT02560753).
Collapse
Affiliation(s)
- Ming Tong
- Liver Research Center, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA.,Division of Gastroenterology, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA.,Department of Medicine, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Chetram Deochand
- Liver Research Center, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA.,Division of Gastroenterology, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA.,Department of Medicine, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - John Didsbury
- T3D Therapeutics, Inc., Research Triangle Park, NC, USA
| | - Suzanne M de la Monte
- Liver Research Center, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA.,Division of Gastroenterology, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA.,Division of Neuropathology, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA.,Department of Medicine, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA.,Department of Pathology, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA.,Department of Neurology, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA.,Department of Neurosurgery, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
50
|
Röszer T. Transcriptional control of apoptotic cell clearance by macrophage nuclear receptors. Apoptosis 2016; 22:284-294. [DOI: 10.1007/s10495-016-1310-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|