1
|
Liu M, Zhao W, Shi R, Wang Z, Li X, Wang D. Analysis of the potential biological mechanisms of geniposide on renal fibrosis by network pharmacology and experimental verification. BMC Pharmacol Toxicol 2025; 26:17. [PMID: 39871379 PMCID: PMC11770925 DOI: 10.1186/s40360-025-00855-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 01/22/2025] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND Renal fibrosis is crucial in the progression of chronic kidney disease (CKD) to end-stage renal failure. Geniposide, an iridoid glycoside, has shown therapeutic potential in acute kidney injury, diabetic nephropathy, and atherosclerosis. The aim of this study was to investigate the role of geniposide in renal fibrosis and its underlying mechanisms. METHODS The network pharmacology and molecular docking methods were used to identify potential targets and pathways of geniposide for treating renal fibrosis. In vivo, the unilateral ureteral obstruction (UUO) mouse model was treated with geniposide. In vitro, TGF-β1-stimulated human renal tubular epithelial (HK-2) cells were applied for validation. HE, PAS, Masson, and immunohistochemistry staining were performed to evaluate its effects on the kidneys of UUO mice. RT-qPCR and western blotting were used to detect the expression of hub genes and signaling pathways. RESULTS 101 overlapping genes were identified, with the top 10 including AKT1, MMP9, GAPDH, BCL2, TNF, CASP3, SRC, EGFR, IL-1β, and STAT1. GO analysis suggested that these key targets were mainly involved in cell proliferation and apoptosis. KEGG analysis revealed that the PI3K/AKT, MAPK, and Rap1 signaling pathways were associated with geniposide against renal fibrosis. Molecular docking suggested a strong binding affinity of geniposide to the hub genes. In vivo experiments showed that geniposide ameliorated kidney injury and fibrosis, and inhibited the mRNA levels of AKT1, MMP9, BCL2, and TNF. In addition, geniposide inhibited the activation of the PI3K/AKT signaling pathway, thereby suppressing renal fibrosis in UUO mice and TGF-β1-induced HK-2 cells. CONCLUSIONS Geniposide can attenuate renal fibrosis by inhibiting the PI3K/AKT pathway, suggesting its potential as a therapeutic agent for renal fibrosis.
Collapse
Affiliation(s)
- Mengqian Liu
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui, 230601, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wenman Zhao
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui, 230601, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Rui Shi
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui, 230601, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhijuan Wang
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui, 230601, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xunliang Li
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui, 230601, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Deguang Wang
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui, 230601, China.
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
2
|
Tetz V, Kardava K, Vecherkovskaya M, Khodadadi-Jamayran A, Tsirigos A, Tetz G. Regulating white blood cell activity through the novel Universal Receptive System. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.06.631232. [PMID: 39896476 PMCID: PMC11785007 DOI: 10.1101/2025.01.06.631232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
The understanding of the mechanisms that control key features of immune cells in various disease contexts remains limited, and few techniques are available for manipulating immune cells. Thus, discovering novel strategies for regulating immune cells is essential for gaining insight into their roles in health and disease. In this study, we investigated the potential of the recently described Universal Receptive System to regulate human immune cell functions. This was achieved for the first time by specifically targeting newly discovered surface-bound DNA and RNA-based receptors on leukocytes and generating "Leukocyte-Tells." This approach upregulated numerous genes related to immune cell signaling, migration, endocytosis, and phagocytosis pathways. The antimicrobial and anticancer activities of Leukocyte-Tells exceeded the activity of control leukocytes in vitro . In some settings, such as in antibiofilm experiments, the Leukocyte-Tells showed up to 1,000,000-fold higher activities than control leukocytes. Our findings reveal, for the first time, that the Universal Receptive System can orchestrate fundamental properties of immune cells, including enhanced antimicrobial and anti-tumor activities. This novel approach offers a new avenue for understanding the biology and regulation of white blood cells.
Collapse
|
3
|
Huang D, Liu J, Yang J, Liang J, Zhang J, Han Q, Yu J, Yang T, Meng Q, Steinberg T, Li C, Chang Z. Restoration of Pregnancy Function Using a GT/PCL Biofilm in a Rabbit Model of Uterine Injury. Tissue Eng Part A 2025; 31:29-44. [PMID: 38526390 DOI: 10.1089/ten.tea.2023.0366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2024] Open
Abstract
Biomaterial scaffolds have been used successfully to promote the regenerative repair of small endometrial lesions in small rodents, providing partial restoration of gestational function. The use of rabbits in this study allowed us to investigate a larger endometrial tissue defect and myometrial injury model. A gelatin/polycaprolactone (GT/PCL) gradient-layer biofilm was sutured at the defect to guide the reconstruction of the original tissue structure. Twenty-eight days postimplantation, the uterine cavity had been restored to its original morphology, endometrial growth was accompanied by the formation of glands and blood vessels, and the fragmented myofibers of the uterine smooth muscle had begun to resemble the normal structure of the lagomorph uterine cavity, arranging in a circular luminal pattern and a longitudinal serosal pattern. In addition, the repair site supported both embryonic implantation into the placenta and normal embryonic development. Four-dimensional label-free proteomic analysis identified the cell adhesion molecules, phagosome, ferroptosis, rap1 signaling pathways, hematopoietic cell lineage, complement and coagulation cascades, tricarboxylic acid cycle, carbon metabolism, and hypoxia inducible factor (HIF)-1 signaling pathways as important in the endogenous repair process of uterine tissue injury, and acetylation of protein modification sites upregulated these signaling pathways.
Collapse
Affiliation(s)
- Di Huang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, P. R. China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, P. R. China
| | - Jing Liu
- The Affiliated Taian City Central Hospital of Qingdao University, Taian, P. R. China
| | - Jie Yang
- The Affiliated Taian City Central Hospital of Qingdao University, Taian, P. R. China
| | - Junhui Liang
- Department of Gynaecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, P. R. China
| | - Jing Zhang
- Shandong First Medical University, Tai'an, P. R. China
| | - Qinyu Han
- Shandong First Medical University, Jinan, P. R. China
| | - Jianlong Yu
- Rizhao People's Hospital, Rizhao, P. R. China
| | - Tingting Yang
- Tai'an Maternal and Child Health Hospital, Tai'an, P. R. China
| | - Qi Meng
- The Affiliated Taian City Central Hospital of Qingdao University, Taian, P. R. China
| | - Thorsten Steinberg
- Division of Oral Biotechnology, Center for Dental Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Changzhong Li
- Center of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, P. R. China
- Institute of Obstetrics and Gynecology, Shenzhen PKU-HKUST Medical Center, Shenzhen, P. R. China
- Shenzhen Key Laboratory on Technology for Early Diagnosis of Major Gynecologic Diseases, Shenzhen, P. R. China
| | - Zhongle Chang
- Shandong Agriculture University, Tai'an, P. R. China
| |
Collapse
|
4
|
Jabłońska M, Sawicki T, Żulewska J, Staniewska K, Łobacz A, Przybyłowicz KE. The Role of Bovine Milk-Derived Exosomes in Human Health and Disease. Molecules 2024; 29:5835. [PMID: 39769923 PMCID: PMC11728725 DOI: 10.3390/molecules29245835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 01/14/2025] Open
Abstract
Bovine milk is widely recognized as one of the most valuable sources of nutrients such as proteins, fats, vitamins, and minerals that support the development and health of the body. In recent years, there has been increasing scientific interest in exosomes, the small membrane-bound vesicles found in milk. Through their content (e.g., microRNA), exosomes can influence gene expression and modulate key signaling pathways within target cells. Results from in vitro and in vivo studies have shown that bovine milk-derived exosomes can alleviate intestinal inflammation by regulating signaling pathways and positively influencing the composition of the gut microbiota. They also improve cognitive function and support nervous system regeneration. In addition, exosomes promote bone health by stimulating osteoblast formation and inhibiting bone resorption, helping to prevent osteoporosis. Studies have shown that exosomes have beneficial effects on skin health by promoting collagen production, protecting cells from oxidative stress, and delaying the ageing process. Bovine milk-derived exosomes are a promising tool for the treatment and prevention of a variety of diseases, particularly those related to inflammation and tissue regeneration. Although these results are promising, further studies are needed to fully understand the mechanisms of action and the potential clinical application of milk exosomes in the prevention and treatment of different diseases.
Collapse
Affiliation(s)
- Monika Jabłońska
- Department of Human Nutrition, Faculty of Food Sciences, University of Warmia and Mazury in Olsztyn, 45f Sloneczna, 10-718 Olsztyn, Poland; (T.S.); (K.E.P.)
| | - Tomasz Sawicki
- Department of Human Nutrition, Faculty of Food Sciences, University of Warmia and Mazury in Olsztyn, 45f Sloneczna, 10-718 Olsztyn, Poland; (T.S.); (K.E.P.)
| | - Justyna Żulewska
- Department of Dairy Science and Quality Management, Faculty of Food Science, University of Warmia and Mazury in Olsztyn, Oczapowskiego 7, 10-719 Olsztyn, Poland; (J.Ż.); (A.Ł.)
| | - Katarzyna Staniewska
- Department of Commodity Science and Food Analysis, Faculty of Food Sciences, University of Warmia and Mazury in Olsztyn, Pl. Cieszynski 1, 10-726 Olsztyn, Poland;
| | - Adriana Łobacz
- Department of Dairy Science and Quality Management, Faculty of Food Science, University of Warmia and Mazury in Olsztyn, Oczapowskiego 7, 10-719 Olsztyn, Poland; (J.Ż.); (A.Ł.)
| | - Katarzyna E. Przybyłowicz
- Department of Human Nutrition, Faculty of Food Sciences, University of Warmia and Mazury in Olsztyn, 45f Sloneczna, 10-718 Olsztyn, Poland; (T.S.); (K.E.P.)
| |
Collapse
|
5
|
Jiang J, Zhang P, Yuan Y, Xu X, Wu T, Zhang Z, Wang J, Bi Y. Prolactin deficiency drives diabetes-associated cognitive dysfunction by inducing microglia-mediated synaptic loss. J Neuroinflammation 2024; 21:295. [PMID: 39543619 PMCID: PMC11566644 DOI: 10.1186/s12974-024-03289-z] [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: 09/18/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND Diabetes-associated cognitive dysfunction, characterized by hippocampal synaptic loss as an early pathological feature, seriously threatens patients' quality of life. Synapses are dynamic structures, and hormones play important roles in modulating the formation and elimination of synapses. The pituitary, the master gland of the body, releases several hormones with multiple roles in hippocampal synaptic regulation. In this study, we aimed to explore the relationship between pituitary hormones and cognitive decline in diabetes. METHODS A total of 744 patients with type 2 diabetes (T2DM) (445 men and 299 postmenopausal women) who underwent serum pituitary hormone level assessments, comprehensive cognitive evaluations and MRI scans were enrolled. Dynamic diet interventions were applied in both chow diet-fed mice and high-fat diet (HFD)-fed diabetic mice. The cognitive performance and hippocampal pathology of prolactin (PRL)-knockout mice, neuronal prolactin receptor (PRLR)-specific knockout mice and microglial PRLR-specific knockout mice were assessed. Microglial PRLR-specific knockout mice were fed an HFD to model diabetes. Diabetic mice received an intracerebroventricular infusion of recombinant PRL protein or vehicle. RESULTS This clinical study revealed that decreased PRL levels were associated with cognitive impairment and hippocampal damage in T2DM patients. In diabetic mice, PRL levels diminished before hippocampal synaptic loss and cognitive decline occurred. PRL loss could directly cause cognitive dysfunction and decreased hippocampal synaptic density. Knockout of PRLR in microglia, rather than neurons, induced hippocampal synaptic loss and cognitive impairment. Furthermore, blockade of PRL/PRLR signaling in microglia exacerbated abnormal microglial phagocytosis of synapses, further aggravating hippocampal synaptic loss and cognitive impairment in diabetic mice. Moreover, PRL infusion reduced microglia-mediated synaptic loss, thereby alleviating cognitive impairment in diabetic mice. CONCLUSION PRL is associated with cognitive dysfunction and hippocampal damage in T2DM patients. In diabetes, a decrease in PRL level drives hippocampal synaptic loss and cognitive impairment by increasing microglia-mediated synapse engulfment. Restoration of PRL levels ameliorates cognitive dysfunction and hippocampal synaptic loss in diabetic mice.
Collapse
Affiliation(s)
- Jiaxuan Jiang
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China
- Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| | - Pengzi Zhang
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China
- Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| | - Yue Yuan
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China
- Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| | - Xiang Xu
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China
- Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| | - Tianyu Wu
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China
- Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| | - Zhou Zhang
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China.
- Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China.
| | - Jin Wang
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China.
- Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China.
| | - Yan Bi
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China.
- Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China.
| |
Collapse
|
6
|
Williams MD, Morgan JS, Bullock MT, Poovey CE, Wisniewski ME, Francisco JT, Barajas-Nunez JA, Hijazi AM, Theobald D, Sriramula S, Mansfield KD, Holland NA, Tulis DA. pH-sensing GPR68 inhibits vascular smooth muscle cell proliferation through Rap1A. Am J Physiol Heart Circ Physiol 2024; 327:H1210-H1229. [PMID: 39269448 PMCID: PMC11560072 DOI: 10.1152/ajpheart.00413.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/06/2024] [Accepted: 09/06/2024] [Indexed: 09/15/2024]
Abstract
Phenotypic transformation of vascular smooth muscle (VSM) from a contractile state to a synthetic, proliferative state is a hallmark of cardiovascular disease (CVD). In CVD, diseased tissue often becomes acidic from altered cellular metabolism secondary to compromised blood flow, yet the contribution of local acid/base imbalance to the disease process has been historically overlooked. In this study, we examined the regulatory impact of the pH-sensing G protein-coupled receptor GPR68 on vascular smooth muscle (VSM) proliferation in vivo and in vitro in wild-type (WT) and GPR68 knockout (KO) male and female mice. Arterial injury reduced GPR68 expression in WT vessels and exaggerated medial wall remodeling in GPR68 KO vessels. In vitro, KO VSM cells showed increased cell-cycle progression and proliferation compared with WT VSM cells, and GPR68-inducing acidic exposure reduced proliferation in WT cells. mRNA and protein expression analyses revealed increased Rap1A in KO cells compared with WT cells, and RNA silencing of Rap1A reduced KO VSM cell proliferation. In sum, these findings support a growth-inhibitory capacity of pH-sensing GPR68 and suggest a mechanistic role for the small GTPase Rap1A in GPR68-mediated VSM growth control. These results shed light on GPR68 and its effector Rap1A as potential targets to combat pathological phenotypic switching and proliferation in VSM.NEW & NOTEWORTHY Extracellular acidosis remains an understudied feature of many pathologies. We examined a potential regulatory role for pH-sensing GPR68 in vascular smooth muscle (VSM) growth in the context of CVD. With in vivo and in vitro growth models with GPR68-deficient mice and GPR68 induction strategies, novel findings revealed capacity of GPR68 to attenuate growth through the small GTPase Rap1A. These observations highlight GPR68 and its effector Rap1A as possible therapeutic targets to combat pathological VSM growth.
Collapse
MESH Headings
- Animals
- Female
- Male
- Mice
- Cell Proliferation
- Cells, Cultured
- Disease Models, Animal
- Hydrogen-Ion Concentration
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- rap1 GTP-Binding Proteins/metabolism
- rap1 GTP-Binding Proteins/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/genetics
- Signal Transduction
- Vascular Remodeling
Collapse
Affiliation(s)
- Madison D Williams
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States
| | - Joshua S Morgan
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States
| | - Michael T Bullock
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States
| | - Cere E Poovey
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States
| | - Michael E Wisniewski
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States
| | - Jake T Francisco
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States
| | - Jerry A Barajas-Nunez
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States
| | - Amira M Hijazi
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States
| | - Drew Theobald
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States
| | - Srinivas Sriramula
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States
| | - Kyle D Mansfield
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States
| | - Nathan A Holland
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States
| | - David A Tulis
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States
| |
Collapse
|
7
|
de Vasconcelos PC, Freitas TR, de Araújo Lopes LV, Peixoto LR, Xavier MP, Cançado Figueiredo AC, Dias KL, de Oliveira JG, de Oliveira Salles PG, Vago AR, de Paula Sabino A, de Lima Rocha MG. RAP1-GTPase immunostaining is altered in human precancerous and cancerous cervical lesions. Biomark Med 2024; 18:771-785. [PMID: 39254347 PMCID: PMC11457648 DOI: 10.1080/17520363.2024.2394384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 08/09/2024] [Indexed: 09/11/2024] Open
Abstract
Aim: This study investigated RAP1 immunostaining variation in different cell types during CC progression.Methods: Paraffin-embedded cervical tissues from 101 patients were categorized into control, pre-neoplastic and neoplastic groups. RAP1 immunolocalization, HPV detection and genotyping were performed. A semiquantitative immunoreactive score was employed to compare labeling intensity, cellular localization, nuclear labeling, percentage and distribution of reactive cells.Results: 73% (72/99) of cervical specimens were HPV+. RAP1 was localized in the nucleus and cytoplasm of all samples. Cytoplasmic RAP1 immunoscore was higher than nuclear score in all CC groups. RAP1 intensity increased with lesion severity. SCC samples exhibited predominantly intense RAP1 immunostaining.Conclusion: RAP1 is an efficient biomarker for detecting invasive CC lesions but has limited utility in distinguishing SCC grades.
Collapse
Affiliation(s)
- Paula Cristina de Vasconcelos
- Department of Clinical & Toxicological Analysis, College of Pharmacy, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Tulio Resende Freitas
- Department of Clinical & Toxicological Analysis, College of Pharmacy, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | | | - Larissa Rodrigues Peixoto
- Department of Morphology, Institute of Biological Sciences, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Marcelo Pascoal Xavier
- Department of Pathological Anatomy, College of Medicine – Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, 30130-100, Brazil
| | - Anna Carolina Cançado Figueiredo
- Integrated Research Group on Biomarkers, Renée Rachou Research Institute – FIOCRUZ, Belo Horizonte, Minas Gerais, 30190-003, Brazil
| | - Karolina Lopes Dias
- Laboratory of Cellular & Molecular Immunology, Renée Rachou Research Institute – FIOCRUZ, Belo Horizonte, Minas Gerais, 30190-003, Brazil
| | - Jaqueline Germano de Oliveira
- Laboratory of Cellular & Molecular Immunology, Renée Rachou Research Institute – FIOCRUZ, Belo Horizonte, Minas Gerais, 30190-003, Brazil
| | | | - Annamaria Ravaro Vago
- Department of Morphology, Institute of Biological Sciences, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Adriano de Paula Sabino
- Department of Clinical & Toxicological Analysis, College of Pharmacy, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Maria Gabrielle de Lima Rocha
- Department of Clinical & Toxicological Analysis, College of Pharmacy, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| |
Collapse
|
8
|
Zisser L, Binder CJ. Extracellular Vesicles as Mediators in Atherosclerotic Cardiovascular Disease. J Lipid Atheroscler 2024; 13:232-261. [PMID: 39355407 PMCID: PMC11439751 DOI: 10.12997/jla.2024.13.3.232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/12/2024] [Accepted: 07/26/2024] [Indexed: 10/03/2024] Open
Abstract
Atherosclerosis is a chronic inflammatory disease of the arterial intima, characterized by accumulation of lipoproteins and accompanying inflammation, leading to the formation of plaques that eventually trigger occlusive thrombotic events, such as myocardial infarction and ischemic stroke. Although many aspects of plaque development have been elucidated, the role of extracellular vesicles (EVs), which are lipid bilayer-delimited vesicles released by cells as mediators of intercellular communication, has only recently come into focus of atherosclerosis research. EVs comprise several subtypes that may be differentiated by their size, mode of biogenesis, or surface marker expression and cargo. The functional effects of EVs in atherosclerosis depend on their cellular origin and the specific pathophysiological context. EVs have been suggested to play a role in all stages of plaque formation. In this review, we highlight the known mechanisms by which EVs modulate atherogenesis and outline current limitations and challenges in the field.
Collapse
Affiliation(s)
- Lucia Zisser
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
9
|
María Del Rocío PB, Palomares Bralo M, Vanhooydonck M, Hamerlinck L, D'haene E, Leimbacher S, Jacobs EZ, De Cock L, D'haenens E, Dheedene A, Malfait Z, Vantomme L, Silva A, Rooney K, Santos-Simarro F, Lleuger-Pujol R, García-Miñaúr S, Losantos-García I, Menten B, Gestri G, Ragge N, Sadikovic B, Bogaert E, Syx D, Callewaert B, Vergult S. Loss-of-function of the Zinc Finger Homeobox 4 ( ZFHX4) gene underlies a neurodevelopmental disorder. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.07.24311381. [PMID: 39148819 PMCID: PMC11326360 DOI: 10.1101/2024.08.07.24311381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
8q21.11 microdeletions encompassing the gene encoding transcription factor ZFHX4, have previously been associated by us with a syndromic form of intellectual disability, hypotonia, decreased balance and hearing loss. Here, we report on 57 individuals, 52 probands and 5 affected family members, with protein truncating variants (n=36), (micro)deletions (n=20) or an inversion (n=1) affecting ZFHX4 with variable developmental delay and intellectual disability, distinctive facial characteristics, morphological abnormalities of the central nervous system, behavioral alterations, short stature, hypotonia, and occasionally cleft palate and anterior segment dysgenesis. The phenotypes associated with 8q21.11 microdeletions and ZFHX4 intragenic loss-of-function variants largely overlap, identifying ZFHX4 as the main driver for the microdeletion syndrome, although leukocyte-derived DNA shows a mild common methylation profile for (micro)deletions only. We identify ZFHX4 as a transcription factor that is increasingly expressed during human brain development and neuronal differentiation. Furthermore, ZFHX4 interacting factors identified via IP-MS in neural progenitor cells, suggest an important role for ZFHX4 in cellular and developmental pathways, especially during histone modifications, cytosolic transport and development. Additionally, using CUT&RUN, we observed that ZFHX4 binds with the promoter regions of genes with crucial roles in embryonic, neuron and axon development. Since loss-of-function variants in ZFHX4 are found with consistent dysmorphic facial features, we investigated whether the disruption of zfhx4 causes craniofacial abnormalities in zebrafish. First-generation (F0) zfhx4 crispant zebrafish, (mosaic) mutant for zfhx4 loss-of-function variants, have significantly shorter Meckel's cartilages and smaller ethmoid plates compared to control zebrafish. Furthermore, behavioral assays show a decreased movement frequency in the zfhx4 crispant zebrafish in comparison with control zebrafish larvae. Although further research is needed, our in vivo work suggests a role for zfhx4 in facial skeleton patterning, palatal development and behavior.
Collapse
Affiliation(s)
- Pérez Baca María Del Rocío
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - María Palomares Bralo
- CIBERER-ISCIII and INGEMM, Institute of Medical and Molecular Genetics, Hospital Universitario La Paz, Madrid, Spain
- ITHACA- European Reference Network, Spain
| | - Michiel Vanhooydonck
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Lisa Hamerlinck
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Eva D'haene
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Sebastian Leimbacher
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Eva Z Jacobs
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Laurenz De Cock
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Erika D'haenens
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Annelies Dheedene
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Zoë Malfait
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Lies Vantomme
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Ananilia Silva
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
| | - Kathleen Rooney
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London, ON, Canada
| | - Fernando Santos-Simarro
- Unit of Molecular Diagnostics and Clinical Genetics, Hospital Universitari Son Espases, Health Research Institute of the Balearic Islands (IdiSBa), Palma, Spain
| | - Roser Lleuger-Pujol
- Hereditary Cancer Program, Catalan Institute of Oncology, Doctor Josep Trueta University Hospital; Precision Oncology Group (OncoGIR-Pro), Institut d'Investigació Biomèdica de Girona (IDIGBI), Girona, Spain
| | - Sixto García-Miñaúr
- CIBERER-ISCIII and INGEMM, Institute of Medical and Molecular Genetics, Hospital Universitario La Paz, Madrid, Spain
- ITHACA- European Reference Network, Spain
| | | | - Björn Menten
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Gaia Gestri
- University College London, London, England, Great Britain
| | - Nicola Ragge
- Birmingham Women's and Children's NHS Foundation Trust, Clinical Genetics Unit, Birmingham Womens Hospital, Lavender House, Mindelsohn Way, Edgbaston, Birmingham B15 2TG
| | - Bekim Sadikovic
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London, ON, Canada
| | - Elke Bogaert
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Delfien Syx
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Bert Callewaert
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Sarah Vergult
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| |
Collapse
|
10
|
Yu J, Du Y, Liu C, Xie Y, Yuan M, Shan M, Li N, Liu C, Wang Y, Qin J. Low GPR81 in ER + breast cancer cells drives tamoxifen resistance through inducing PPARα-mediated fatty acid oxidation. Life Sci 2024; 350:122763. [PMID: 38823505 DOI: 10.1016/j.lfs.2024.122763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/13/2024] [Accepted: 05/28/2024] [Indexed: 06/03/2024]
Abstract
AIMS The intricate molecular mechanisms underlying estrogen receptor-positive (ER+) breast carcinogenesis and resistance to endocrine therapy remain elusive. In this study, we elucidate the pivotal role of GPR81, a G protein-coupled receptor, in ER+ breast cancer (BC) by demonstrating low expression of GPR81 in tamoxifen (TAM)-resistant ER+ BC cell lines and tumor samples, along with the underlying molecular mechanisms. MAIN METHODS Fatty acid oxidation (FAO) levels and lipid accumulation were explored using MDA and FAβO assay, BODIPY 493/503 staining, and Lipid TOX staining. Autophagy levels were assayed using CYTO-ID detection and Western blotting. The impact of GPR81 on TAM resistance in BC was investigated through CCK8 assay, colony formation assay and a xenograft mice model. RESULTS Aberrantly low GPR81 expression in TAM-resistant BC cells disrupts the Rap1 pathway, leading to the upregulation of PPARα and CPT1. This elevation in PPARα/CPT1 enhances FAO, impedes lipid accumulation and lipid droplet (LD) formation, and subsequently inhibits cell autophagy, ultimately promoting TAM-resistant BC cell growth. Moreover, targeting GPR81 and FAO emerges as a promising therapeutic strategy, as the GPR81 agonist and the CPT1 inhibitor etomoxir effectively inhibit ER+ BC cell and tumor growth in vivo, re-sensitizing TAM-resistant ER+ cells to TAM treatment. CONCLUSION Our data highlight the critical and functionally significant role of GPR81 in promoting ER+ breast tumorigenesis and resistance to endocrine therapy. GPR81 and FAO levels show potential as diagnostic biomarkers and therapeutic targets in clinical settings for TAM-resistant ER+ BC.
Collapse
Affiliation(s)
- Jing Yu
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Yongjun Du
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Chang Liu
- School of Medicine, Nankai University, Tianjin 300071, China.
| | - Yu Xie
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Mengci Yuan
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Meihua Shan
- Department of Clinical Biochemistry, Army Medical University, Chongqing 400038, China
| | - Ning Li
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China
| | - Chang Liu
- School of Medicine, Nankai University, Tianjin 300071, China.
| | - Yue Wang
- School of Medicine, Nankai University, Tianjin 300071, China; Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Hospital of Stomatology, Nankai University, Tianjin 300041, China.
| | - Junfang Qin
- School of Medicine, Nankai University, Tianjin 300071, China.
| |
Collapse
|
11
|
Khan I, Muneer R, Qazi REM, Salim A. Pharmacological activation of mesenchymal stem cells increases gene expression pattern of cell adhesion molecules and fusion with neonatal cardiomyocytes. Cell Biochem Funct 2024; 42:e4090. [PMID: 38973147 DOI: 10.1002/cbf.4090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 06/27/2024] [Accepted: 06/30/2024] [Indexed: 07/09/2024]
Abstract
Cellular therapy is considered a better option for the treatment of degenerative disorders. Different cell types are being used for tissue regeneration. Despite extensive research in this field, several issues remain to be addressed concerning cell transplantation. One of these issues is the survival and homing of administered cells in the injured tissue, which depends on the ability of these cells to adhere. To enhance cell adherence and survival, Rap1 GTPase was activated in mesenchymal stem cells (MSCs) as well as in cardiomyocytes (CMs) by using 8-pCPT-2'-O-Me-cAMP, and the effect on gene expression dynamics was determined through quantitative reverse transcriptase-polymerase chain reaction analysis. Pharmacological activation of MSCs and CMs resulted in the upregulation of connexin-43 and cell adhesion genes, which increased the cell adhesion ability of MSCs and CMs, and increased the fusion of MSCs with neonatal CMs. Treating stem cells with a pharmacological agent that activates Rap1a before transplantation can enhance their fusion with CMs and increase cellular regeneration.
Collapse
Affiliation(s)
- Irfan Khan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
- Center for Regenerative Medicine and Stem Cell Research, The Aga Khan University, Karachi, Pakistan
- Department of Ophthalmology and Visual Sciences, The Aga Khan University, Karachi, Pakistan
- Department of Biological and Biomedical Sciences, The Aga Khan University, Karachi, Pakistan
| | - Rabbia Muneer
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Rida-E-Maria Qazi
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
- Center for Regenerative Medicine and Stem Cell Research, The Aga Khan University, Karachi, Pakistan
| | - Asmat Salim
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| |
Collapse
|
12
|
Shi M. The Efficacy of Ganoderma lucidum Extracts on Treating Endometrial Cancer: A Network Pharmacology Approach. Reprod Sci 2024; 31:1881-1894. [PMID: 38448739 PMCID: PMC11217070 DOI: 10.1007/s43032-024-01500-3] [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: 07/28/2023] [Accepted: 02/09/2024] [Indexed: 03/08/2024]
Abstract
Ganoderma lucidum (GL) is a prominent medicinal mushroom in traditional Chinese medicine, known for its potent antitumor properties. This study aimed to illustrate the efficacy of GL extracts (GLE) on treating endometrial cancer (EC) and explore the underlying mechanisms via network pharmacology and experimental validation. Network pharmacological analysis was conducted to explore the therapeutic efficacy and mechanisms of GL on EC. In vitro experimental validation was performed on human endometrial cancer cell lines HEC-1-A and KLE. Network pharmacology revealed that key targets of GL against EC were primarily associated with the Rap1 signaling pathway. In in vitro experiments, GLE or GGTI-298 (a GTPase inhibitor) treatment inhibited cell proliferation and migration, promoted cell apoptosis, increased caspase-3 level, and arrested cell cycle in G1 phase in HEC-1-A and KLE cells. GLE increased the protein expression of Rap1-GTP, p-AKT, and p-ERK2 in HEC-1-A and KLE cells. Moreover, GGTI-298 enhanced the effects of GLE on suppressing the malignant progression of EC cells and on activating Rap1 signaling pathway. GLE inhibited the malignant progression of EC cells probably via activating the Rap1 signaling pathway.
Collapse
Affiliation(s)
- Min Shi
- Department of Medical Oncology, Zhejiang Putuo Hospital, Zhoushan, 316100, Zhejiang Province, China.
| |
Collapse
|
13
|
Zhang H, Yang Y, Cao Y, Guan J. Effects of chronic stress on cancer development and the therapeutic prospects of adrenergic signaling regulation. Biomed Pharmacother 2024; 175:116609. [PMID: 38678960 DOI: 10.1016/j.biopha.2024.116609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/14/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024] Open
Abstract
Long-term chronic stress is an important factor in the poor prognosis of cancer patients. Chronic stress reduces the tissue infiltration of immune cells in the tumor microenvironment (TME) by continuously activating the adrenergic signaling, inhibits antitumor immune response and tumor cell apoptosis while also inducing epithelial-mesenchymal transition (EMT) and tumor angiogenesis, promoting tumor invasion and metastasis. This review first summarizes how adrenergic signaling activates intracellular signaling by binding different adrenergic receptor (AR) heterodimers. Then, we focused on reviewing adrenergic signaling to regulate multiple functions of immune cells, including cell differentiation, migration, and cytokine secretion. In addition, the article discusses the mechanisms by which adrenergic signaling exerts pro-tumorigenic effects by acting directly on the tumor itself. It also highlights the use of adrenergic receptor modulators in cancer therapy, with particular emphasis on their potential role in immunotherapy. Finally, the article reviews the beneficial effects of stress intervention measures on cancer treatment. We think that enhancing the body's antitumor response by adjusting adrenergic signaling can enhance the efficacy of cancer treatment.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Oncology, The Eighth Medical Center, Chinese PLA (People's Liberation Army) General Hospital, Beijing 100091, China; Department of Oncology, The Fifth Medical Center, Chinese PLA (People's Liberation Army) General Hospital, Beijing 100071, China.
| | - Yuwei Yang
- College of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing Key Laboratory of OTIR, Beijing, 100091, China.
| | - Yan Cao
- College of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing Key Laboratory of OTIR, Beijing, 100091, China.
| | - Jingzhi Guan
- Department of Oncology, The Fifth Medical Center, Chinese PLA (People's Liberation Army) General Hospital, Beijing 100071, China.
| |
Collapse
|
14
|
Erban T, Markovic M, Sopko B. Sublethal acetamiprid exposure induces immunity, suppresses pathways linked to juvenile hormone synthesis in queens and affects cycle-related signaling in emerging bees. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 349:123901. [PMID: 38556147 DOI: 10.1016/j.envpol.2024.123901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 03/05/2024] [Accepted: 03/25/2024] [Indexed: 04/02/2024]
Abstract
Acetamiprid is the only neonicotinoid registered in the European Union because the risks of neonicotinoids to honey bees and other pollinators are strictly regulated. Herein, we orally exposed honey bee colonies to sublethal concentrations of acetamiprid (20 μg/L) under isolated conditions. After one month of continuous exposure, the emerging bees and queens were collected and analyzed via high-throughput label-free quantitative proteomics using a data-independent acquisition strategy. Six and 34 significantly differentially expressed proteins (DEPs) were identified in the emerging bees and queens, respectively. Mrjp3 was the only DEP found in both sample types/castes, and its opposite regulation illustrated a differential response. The DEPs in the emerging bees (H/ACA RNP, Rap1GAP, Mrjp3, and JHE) suggested that sublethal exposure to acetamiprid affected cell cycle-related signaling, which may affect the life history of workers in the colony. The DEPs with increased levels in queens, such as Mrjps 1-4 and 6-7, hymenoptaecin, and apidaecin 22, indicated an activated immune response. Additionally, the level of farnesyl pyrophosphate synthase (FPPS), which is essential for the mevalonate pathway and juvenile hormone biosynthesis, was significantly decreased in queens. The impaired utilization of juvenile hormone in queens supported the identification of additional DEPs. Furthermore, the proteome changes suggested the existence of increased neonicotinoid detoxification by UDP-glucuronosyltransferase and increased amino acid metabolism. The results suggest that the continuous exposure of bee colonies to acetamiprid at low doses (nanograms per gram in feed) may pose a threat to the colonies. The different exposure routes and durations for the emerging bees and queens in our experiment must be considered, i.e., the emerging bees were exposed as larvae via feeding royal jelly and beebread provided by workers (nurse bees), whereas the queens were fed royal jelly throughout the experiment. The biological consequences of the proteomic changes resulting from sublethal/chronic exposure require future determination.
Collapse
Affiliation(s)
- Tomas Erban
- Crop Research Institute, Drnovska 507/73, Prague 6-Ruzyne, CZ-161 06, Czechia.
| | - Martin Markovic
- Crop Research Institute, Drnovska 507/73, Prague 6-Ruzyne, CZ-161 06, Czechia
| | - Bruno Sopko
- Crop Research Institute, Drnovska 507/73, Prague 6-Ruzyne, CZ-161 06, Czechia
| |
Collapse
|
15
|
Fisher TW, Munyaneza JE, Brown JK. Sub-optimal temperatures lead to altered expression of stress-related genes and increased 'C andidatus Liberibacter solanacearum' accumulation in potato psyllid. FRONTIERS IN INSECT SCIENCE 2024; 3:1279365. [PMID: 38469510 PMCID: PMC10926459 DOI: 10.3389/finsc.2023.1279365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 11/23/2023] [Indexed: 03/13/2024]
Abstract
Introduction The potato psyllid Bactericera cockerelli is the insect vector of the fastidious bacterium 'Candidatus Liberibacter solanacearum'. The bacterium infects both B. cockerelli and plant species, causing zebra chip (ZC) disease of potato and vein-greening disease of tomato. Temperatures are known to influence the initiation and progression of disease symptom in the host plant, and seasonal transitions from moderate to high temperatures trigger psyllid dispersal migration to facilitate survival. Methods 'Ca. L. solanacearum' -infected and uninfected psyllids were reared at previously established 'permissible', optimal, and 'non-permissible' and temperatures of 18°C, 24°C, and 30°C, respectively. Gene expression profiles for 'Ca. L. solanacearum'-infected and -uninfected adult psyllids reared at different temperatures were characterized by Illumina RNA-Seq analysis. Bacterial genome copy number was quantified by real-time quantitative-PCR (qPCR) amplification. Results Relative gene expression profiles varied in psyllids reared at the three experimental temperatures. Psyllids reared at 18°C and 30°C exhibited greater fold-change increased expression of stress- and 'Ca. L. solanacearum' invasion-related proteins. Quantification by qPCR of bacterial genome copy number revealed that 'Ca. L. solanacearum' accumulation was significantly lower in psyllids reared at 18°C and 30°C, compared to 24°C. Discussion Temperature is a key factor in the life history of potato psyllid and multiplication/accumulation of 'Ca. L. solanacearum' in both the plant and psyllid host, influences the expression of genes associated with thermal stress tolerance, among others, and may have been instrumental in driving the co-evolution of the pathosystem.
Collapse
Affiliation(s)
- Tonja W. Fisher
- School of Plant Sciences, The University of Arizona, Tucson, AZ, United States
| | - Joseph E. Munyaneza
- United States Department of Agriculture-Agricultural Research Service (USDA-ARS), Yakima Agricultural Research Laboratory (YARL), Wapato, WA, United States
| | - Judith K. Brown
- School of Plant Sciences University of Arizona, Tucson, AZ, United States
| |
Collapse
|
16
|
Agarwal H, Tinsley B, Sarecha AK, Ozcan L. Rap1 in the Context of PCSK9, Atherosclerosis, and Diabetes. Curr Atheroscler Rep 2023; 25:931-937. [PMID: 37979063 DOI: 10.1007/s11883-023-01162-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2023] [Indexed: 11/19/2023]
Abstract
PURPOSE OF REVIEW The focus of this article is to highlight the importance of the small GTPase, Ras-associated protein 1 (Rap1), in proprotein convertase subtilisin/kexin type 9 (PCSK9) regulation and atherosclerosis and type 2 diabetes etiology and discuss the potential therapeutic implications of targeting Rap1 in these disease areas. REVIEW FINDINGS Cardiometabolic disease characterized by obesity, glucose intolerance, dyslipidemia, and atherosclerotic cardiovascular disease remain an important cause of mortality. Evidence using mouse models of obesity and insulin resistance indicates that Rap1 deficiency increases proatherogenic PCSK9 and low-density lipoprotein cholesterol levels and predisposes these mice to develop obesity- and statin-induced hyperglycemia, which highlights Rap1's role in cardiometabolic dysfunction. Rap1 may also contribute to cardiovascular disease through its effects on vascular wall cells involved in the atherosclerosis progression. Rap1 activation, specifically in the liver, could be beneficial in the prevention of cardiometabolic perturbations, including type 2 diabetes, hypercholesterolemia, and atherosclerosis.
Collapse
Affiliation(s)
- Heena Agarwal
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Brea Tinsley
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Amesh K Sarecha
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Lale Ozcan
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
17
|
Gao H, Kuang Y, Liu Y, Zhang Y, Wang P, Ma Q. Changes of plasma Rap1A levels in patients with in-stent restenosis after percutaneous coronary intervention and the underlying mechanisms. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2023; 48:1650-1658. [PMID: 38432855 PMCID: PMC10929945 DOI: 10.11817/j.issn.1672-7347.2023.230285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Indexed: 03/05/2024]
Abstract
OBJECTIVES Percutaneous coronary intervention (PCI) is one of the most important treatments for coronary artery disease (CAD). However, in-stent restenosis (ISR) after PCI is a serious complication without effective measures for prevention and treatment. This study aims to investigate the Ras-related protein 1A (Rap1A) level in ISR patients and in the tumor necrosis factor-α (TNF-α)-induced inflammatory injury model of human umbilical vein endothelial cells (HUVECs), to explore the role of Rap1A in regulating TNF-α-induced inflammation in HUVECs and to provide a new potential target for ISR prevention and treatment. METHODS A total of 60 CAD patients, who underwent PCI between December 2020 and July 2022 from the Department of Cardiovascular Medicine of Xiangya Hospital, Central South University, and re-examined coronary angiography (CAG) 1 year after the operation, were included. After admission, 27 patients were diagnosed with ISR and 33 patients were diagnosed with non-in-stent restenosis (non-ISR) according to the CAG. Clinical data were collected, and the plasma Rap1A level was determined by enzyme linked immunosorbent assay (ELISA). In cell experiments, an inflammatory injury model was established with TNF-α treatment (10 ng/mL, 24 h) in HUVECs. The mRNA and protein expression levels of Rap1A, interlukin-6 (IL-6), and vascular cell adhesion molecule-1 (VCAM-1) were measured by real-time reverse transcription PCR and Western blotting. Small interfering RNA (siRNA) was used to explore the role of Rap1A in regulating TNF-α-induced inflammation in HUVECs. RESULTS Compared with the non-ISR patients, a higher proportion of ISR patients had a history of smoking (P=0.005) and diabetes (P=0.028), and higher levels of glycosylated hemoglobin (HbA1c) (P=0.012), low-density lipoprotein cholesterol (LDL-c) (P=0.014), and hypersensitive C-reactive protein (hs-CRP) (P=0.027). The remaining projects did not show significant differences (all P>0.05). The plasma level of Rap1A in the ISR group was significantly higher than that in the non-ISR group [942.14 (873.28 to 1 133.81) μg/mL vs 886.93 (812.61 to 930.98) μg/mL; P=0.004]. Diabetes, LDL-c, and Rap1A were risk factors for ISR by univariate logistic regression analysis (all P<0.05). The mRNA and protein expression levels of inflammatory factors IL-6 and VCAM-1 were increased in HUVECs after 10 ng/mL TNF-α treatment for 24 h compared with the control group (all P<0.05), while the mRNA and protein levels of Rap1A were increased (both P<0.05). After inhibition of Rap1A in HUVECs, the mRNA and protein expression levels of IL-6 and VCAM-1 were significantly decreased (all P<0.05). CONCLUSIONS The plasma Rap1A level was significantly elevated in patients with ISR, suggesting that Rap1A may be a potential biomarker for predicting ISR. In the TNF-α- induced HUVECs inflammatory injury model, the expression level of Rap1A was increased. The level of TNF-α-induced endothelial cell inflammation was decreased after inhibition of Rap1A expression, suggesting that Rap1A may be a potential target for the treatment of endothelial cell inflammation in ISR.
Collapse
Affiliation(s)
- Haodong Gao
- Department of Cardiology, Xiangya Hospital, Central South University; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha 410008.
| | - Yuanyuan Kuang
- Department of Cardiology, Xiangya Hospital, Central South University; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha 410008
| | - Yubo Liu
- Department of Cardiology, Xiangya Hospital, Central South University; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha 410008
| | - Yinzhuang Zhang
- Department of Cardiology, First Hospital of Changsha, Changsha 410005, China
| | - Ping Wang
- Department of Cardiology, Xiangya Hospital, Central South University; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha 410008
| | - Qilin Ma
- Department of Cardiology, Xiangya Hospital, Central South University; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha 410008.
| |
Collapse
|
18
|
Tong J, Li H, Zhang C. Altered 5-methylcytosine modification of mRNA is involved in the pathogenesis of pre-eclampsia. J Cell Biochem 2023; 124:1720-1733. [PMID: 37796115 DOI: 10.1002/jcb.30479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/08/2023] [Accepted: 09/19/2023] [Indexed: 10/06/2023]
Abstract
5-Methylcytosine (m5 C) is a prevalent RNA modification in messenger RNAs (mRNAs). Despite its abundance, its role in the decidua of pre-eclampsia (PE) remains elusive. In this study, we utilized methylated RNA immunoprecipitation sequencing (MeRIP-seq) and RNA-sequencing (RNA-seq) to map m5 C peaks and mRNA expression profile in the decidua of human early-onset PE (EPE), late-onset PE (LPE), and normal pregnancy (NP). Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses elucidated potential roles of the differentially methylated mRNAs (DMGs) and differentially expressed mRNAs in decidualization pathways. Integrative analysis of MeRIP-seq and RNA-seq data pinpointed 50 candidate genes linked to PE, marked by both differentially methylated m5 C peaks and congruent expression changes. To validate these observations, we selected nine genes for verification via quantitative PCR. Our results underscore the precision and reproducibility of our bioinformatics approach. Importantly, we propose that changes in m5 C modification and expression of relevant mRNA might influence the pathogenesis of PE by hampering decidualization. This work shines light on the distinct mRNA m5 C modification patterns and expression profiles in the decidua of PE, implicating pivotal signaling disruptions and decidualization impediments in the onset of PE.
Collapse
Affiliation(s)
- Jing Tong
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Hongwanyu Li
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Cong Zhang
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| |
Collapse
|
19
|
Hiremath C, Gao L, Geshow K, Patterson Q, Barlow H, Cleaver O, Marciano DK. Rap1 regulates lumen continuity via Afadin in renal epithelia. Dev Biol 2023; 501:20-27. [PMID: 37276970 PMCID: PMC10460627 DOI: 10.1016/j.ydbio.2023.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 04/13/2023] [Accepted: 05/11/2023] [Indexed: 06/07/2023]
Abstract
The continuity of a lumen within an epithelial tubule is critical for its function. We previously found that the F-actin binding protein Afadin is required for timely lumen formation and continuity in renal tubules formed from the nephrogenic mesenchyme in mice. Afadin is a known effector and interactor of the small GTPase Rap1, and in the current study, we examine the role of Rap1 in nephron tubulogenesis. Here, we demonstrate that Rap1 is required for nascent lumen formation and continuity in cultured 3D epithelial spheroids and in vivo in murine renal epithelial tubules derived from the nephrogenic mesenchyme, where its absence ultimately leads to severe morphogenetic defects in the tubules. By contrast, Rap1 is not required for lumen continuity or morphogenesis in renal tubules derived from the ureteric epithelium, which differ in that they form by extension from a pre-existing tubule. We further demonstrate that Rap1 is required for correct localization of Afadin to adherens junctions both in vitro and in vivo. Together, these results suggest a model in which Rap1 localizes Afadin to junctional complexes, which in turn regulates nascent lumen formation and positioning to ensure continuous tubulogenesis.
Collapse
Affiliation(s)
- Chitkale Hiremath
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Lei Gao
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Kenya Geshow
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Quinten Patterson
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Haley Barlow
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Ondine Cleaver
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Denise K Marciano
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA.
| |
Collapse
|
20
|
Wei Z, Xia K, Zhou B, Zheng D, Guo W. Zyxin Inhibits the Proliferation, Migration, and Invasion of Osteosarcoma via Rap1-Mediated Inhibition of the MEK/ERK Signaling Pathway. Biomedicines 2023; 11:2314. [PMID: 37626810 PMCID: PMC10452081 DOI: 10.3390/biomedicines11082314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/02/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
Zyxin (ZYX) is an actin-interacting protein with unknown biological functions in patients with osteosarcoma. This research sought to understand how ZYX affects the biological behavior of osteosarcoma cells and to identify the associated mechanism. Firstly, ZYX expression was decreased in osteosarcoma, and its higher expression indicated better outcomes in patients with osteosarcoma. ZYX overexpression significantly inhibited the proliferation, migration, and invasion of osteosarcoma cells, whereas ZYX silencing resulted in the opposite trend. Subsequently, we found that the Rap1 signaling pathway was significantly correlated with ZYX expression as reported in The Cancer Genome Atlas's database using bioinformatic analysis. Moreover, we found that ZYX overexpression regulated the Rap1/MEK/ERK axis, and osteosarcoma cell growth, migration, and invasion were consequently restrained. Additionally, by administering tumor cells subcutaneously to nude mice, a mouse model of transplanted tumors was created. Compared to the control group, the ZYX overexpression group's tumors were lighter and smaller, and the ZYX/Rap1 axis was activated in the ZYX overexpression group. Taken together, our results suggest that ZYX inhibits osteosarcoma cell proliferation, migration, and invasion by regulating the Rap1/MEK/ERK signaling pathway. ZYX might be crucial in the clinical management of osteosarcoma and is a promising novel therapeutic target in patients with this disease.
Collapse
Affiliation(s)
- Zhun Wei
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Kezhou Xia
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Bin Zhou
- Department of Orthopedics, Ezhou Central Hospital, Ezhou 436000, China
| | - Di Zheng
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Weichun Guo
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
21
|
Dehghanian M, Yarahmadi G, Sandoghsaz RS, Khodadadian A, Shamsi F, Vahidi Mehrjardi MY. Evaluation of Rap1GAP and EPAC1 Gene Expression in Endometriosis Disease. Adv Biomed Res 2023; 12:101. [PMID: 37288024 PMCID: PMC10241619 DOI: 10.4103/abr.abr_86_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/30/2022] [Accepted: 07/03/2022] [Indexed: 06/09/2023] Open
Abstract
Background Endometriosis is a female reproductive system disease in which the endometrial tissue is found in other women's organs. Various factors are effective in the development of endometriosis, and because of the interaction of genetics and environmental factors, this disease is a multi-factorial disease. MAPK/ERK and PI3K/Akt/mTOR pathways are activated by growth factors and steroid hormones and are known as two important pathways involved in the processes of growth, proliferation, and survival of endometriosis cells. Raps, monomeric GTPase of the Ras family, are able to activate these pathways independent of Ras. The goal of our study was to evaluate the expression level of Rap1GAP and EPAC1 genes as two important RapGAPs (GTPase-activating proteins) and RapGEFs (guanine nucleotide exchange factors), respectively, in endometriosis tissues and normal endometrium tissues. Materials and Methods In this study, 15 samples of women without signs of endometriosis were taken as control samples. Fifteen ectopic and 15 eutopic samples were taken from women with endometriosis using laparoscopic surgery. The expression of EPAC1 and Rap1GAP genes was investigated by the real-time polymerase chain reaction technique, and the results were analyzed by one-way ANOVA test. Results EPAC1 upregulated significantly in ectopic tissues compared to eutopic and control tissues. Rap1GAP expression was lower in ectopic tissues compared to control and eutopic tissues. Conclusions Based on these results, it may be concluded that changes in the expression of the Rap1GAP and Epca1 genes may play a role in the pathways involved in the pathogenesis, displacement, and migration of endometriosis cells.
Collapse
Affiliation(s)
- Mehran Dehghanian
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Abortion Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Ghafour Yarahmadi
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Abortion Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Reyhaneh Sadat Sandoghsaz
- Abortion Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Ali Khodadadian
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Farimah Shamsi
- Department of Biostatistics and Epidemiology, Center for Healthcare Data Modeling, School of Pulic Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mohammad Yahya Vahidi Mehrjardi
- Research Center for Food Hygiene and Safety, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
22
|
Boi R, Bergwall L, Ebefors K, Bergö MO, Nyström J, Buvall L. Podocyte Geranylgeranyl Transferase Type-I Is Essential for Maintenance of the Glomerular Filtration Barrier. J Am Soc Nephrol 2023; 34:641-655. [PMID: 36735952 PMCID: PMC10103324 DOI: 10.1681/asn.0000000000000062] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 11/25/2022] [Indexed: 01/22/2023] Open
Abstract
SIGNIFICANCE STATEMENT A tightly regulated actin cytoskeleton attained through balanced activity of RhoGTPases is crucial to maintaining podocyte function. However, how RhoGTPases are regulated by geranylgeranylation, a post-translational modification, has been unexplored. The authors found that loss of the geranylgeranylation enzyme geranylgeranyl transferase type-I (GGTase-I) in podocytes led to progressive albuminuria and foot process effacement in podocyte-specific GGTase-I knockout mice. In cultured podocytes, the absence of geranylgeranylation resulted in altered activity of its downstream substrates Rac1, RhoA, Cdc42, and Rap1, leading to alterations of β1-integrins and actin cytoskeleton structural changes. These findings highlight the importance of geranylgeranylation in the dynamic management of RhoGTPases and Rap1 to control podocyte function, providing new knowledge about podocyte biology and glomerular filtration barrier function. BACKGROUND Impairment of the glomerular filtration barrier is in part attributed to podocyte foot process effacement (FPE), entailing disruption of the actin cytoskeleton and the slit diaphragm. Maintenance of the actin cytoskeleton, which contains a complex signaling network through its connections to slit diaphragm and focal adhesion proteins, is thus considered crucial to preserving podocyte structure and function. A dynamic yet tightly regulated cytoskeleton is attained through balanced activity of RhoGTPases. Most RhoGTPases are post-translationally modified by the enzyme geranylgeranyl transferase type-I (GGTase-I). Although geranylgeranylation has been shown to regulate activities of RhoGTPases and RasGTPase Rap1, its significance in podocytes is unknown. METHODS We used immunofluorescence to localize GGTase-I, which was expressed mainly by podocytes in the glomeruli. To define geranylgeranylation's role in podocytes, we generated podocyte-specific GGTase-I knockout mice. We used transmission electron microscopy to evaluate FPE and measurements of urinary albumin excretion to analyze filtration barrier function. Geranylgeranylation's effects on RhoGTPases and Rap1 function were studied in vitro by knockdown or inhibition of GGTase-I. We used immunocytochemistry to study structural modifications of the actin cytoskeleton and β1 integrins. RESULTS Depletion of GGTase-I in podocytes in vivo resulted in FPE and concomitant early-onset progressive albuminuria. A reduction of GGTase-I activity in cultured podocytes disrupted RhoGTPase balance by markedly increasing activity of RhoA, Rac1, and Cdc42 together with Rap1, resulting in dysregulation of the actin cytoskeleton and altered distribution of β1 integrins. CONCLUSIONS These findings indicate that geranylgeranylation is of crucial importance for the maintenance of the delicate equilibrium of RhoGTPases and Rap1 in podocytes and consequently for the maintenance of glomerular integrity and function.
Collapse
Affiliation(s)
- Roberto Boi
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lovisa Bergwall
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kerstin Ebefors
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Martin O. Bergö
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Institute of Biosciences and Nutrition, Karolinska Institute, Solna, Sweden
| | - Jenny Nyström
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lisa Buvall
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Bioscience Renal, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
23
|
Zhu C, Chen Y, Tai Z, Pan H, Shen M, Chen Z, Zhu Q. Effect and mechanism of longkui yinxiao soup in treating psoriasis in mice. Front Pharmacol 2023; 14:1136604. [PMID: 36992832 PMCID: PMC10040526 DOI: 10.3389/fphar.2023.1136604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 02/22/2023] [Indexed: 03/14/2023] Open
Abstract
Objective: Longkui Yinxiao Soup is a traditional Chinese medicine formula used to treat psoriasis for decades. Although Longkui Yinxiao Soup showed promising efficacy in clinical practice, the regulatory mechanisms of Longkui Yinxiao Soup remain elusive. This study aimed to explore the underlying mechanisms of Longkui Yinxiao Soup in a psoriasis-like mouse model.Methods: Longkui Yinxiao Soup was quality controlled by determining the contents of imperatorin and rhoifolin using high-performance liquid chromatography. The imiquimod-induced psoriasis-like mouse model was used to study the therapeutic effect and mechanism of Longkui Yinxiao Soup. The histopathological skin changes were observed by hematoxylin and eosin staining; the infiltration of proliferating proteins, proliferating cell nuclear antigen and Ki67, in skin tissues were observed by immunohistochemical analysis; and the inflammatory factors such as interleukin (IL)-6, tumor necrosis factor (TNF)-α, IL-23, and IL-17 in serum were detected using enzyme-linked immunosorbent assay. RNA sequencing and bioinformatic analysis were used to predict the mechanism of LYS against psoriasis. mRNA expressions of p38, extracellular regulated protein kinases (ERK), mitogen-activated protein kinase 3 (MEK3), mitogen-activated protein kinase 6 (MEK6), RAP1 GTPase activating protein (Rap1gap), and Rap1 were determined using real-time quantitative polymerase chain reaction. The expression levels of proteins related to Rap1–mitogen-activated protein kinase signaling pathways were measured by Western blotting.Results: A quality-control method for Longkui Yinxiao Soup was successfully established using imperatorin and rhoifolin as content determination indexes. Longkui Yinxiao Soup significantly ameliorated the psoriatic symptoms in mice. The serum levels of inflammatory cytokines such as IL-6, TNF-α, IL-23, and IL-17 were decreased, and the expression levels of antigen identified by monoclonal antibody Ki67 (Ki67) and PCNA in skin tissues were downregulated. Moreover, the inhibition of Rap1–MAPK signaling pathways by Longkui Yinxiao Soup was detected.Conclusion: This study confirmed the antipsoriatic activity of Longkui Yinxiao Soup in psoriasis-like mice. This might be due to the inhibition of inflammatory factor secretion, keratinocyte proliferation, and the Rap1–MAPK signal pathway.
Collapse
Affiliation(s)
- Congcong Zhu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, China
| | - Ya Chen
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, China
| | - Zongguang Tai
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, China
| | - Huijun Pan
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, China
| | - Min Shen
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, China
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, China
- *Correspondence: Quangang Zhu, ; Zhongjian Chen,
| | - Quangang Zhu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, China
- *Correspondence: Quangang Zhu, ; Zhongjian Chen,
| |
Collapse
|
24
|
Ferri G, Musikant D, Edreira MM. Host Cell Rap1b mediates cAMP-dependent invasion by Trypanosoma cruzi. PLoS Negl Trop Dis 2023; 17:e0011191. [PMID: 36897926 PMCID: PMC10032529 DOI: 10.1371/journal.pntd.0011191] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 03/22/2023] [Accepted: 02/22/2023] [Indexed: 03/11/2023] Open
Abstract
Trypanosoma cruzi cAMP-mediated invasion has long been described, however, the detailed mechanism of action of the pathway activated by this cyclic nucleotide still remains unknown. We have recently demonstrated a crucial role for Epac in the cAMP-mediated invasion of the host cell. In this work, we gathered evidence indicating that the cAMP/Epac pathway is activated in different cells lines. In accordance, data collected from pull-down experiments designed to identify only the active form of Rap1b (Rap1b-GTP), and infection assays using cells transfected with a constitutively active mutant of Rap1b (Rap1b-G12V), strongly suggest the participation of Rap1b as mediator of the pathway. In addition to the activation of this small GTPase, fluorescence microscopy allowed us to demonstrate the relocalization of Rap1b to the entry site of the parasite. Moreover, phospho-mimetic and non-phosphorylable mutants of Rap1b were used to demonstrate a PKA-dependent antagonistic effect on the pathway, by phosphorylation of Rap1b, and potentially of Epac. Finally, Western Blot analysis was used to determine the involvement of the MEK/ERK signalling downstream of cAMP/Epac/Rap1b-mediated invasion.
Collapse
Affiliation(s)
- Gabriel Ferri
- CONICET-Universidad de Buenos Aires, IQUIBICEN, Ciudad de Buenos Aires, Argentina
- Laboratorio de Biología Molecular de Trypanosomas, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos, Ciudad de Buenos Aires, Argentina
| | - Daniel Musikant
- CONICET-Universidad de Buenos Aires, IQUIBICEN, Ciudad de Buenos Aires, Argentina
- Laboratorio de Biología Molecular de Trypanosomas, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos, Ciudad de Buenos Aires, Argentina
| | - Martin M Edreira
- CONICET-Universidad de Buenos Aires, IQUIBICEN, Ciudad de Buenos Aires, Argentina
- Laboratorio de Biología Molecular de Trypanosomas, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos, Ciudad de Buenos Aires, Argentina
| |
Collapse
|
25
|
Chuan W, Yuan L, Wen J, Jianwei Z, Caiji W, Zeqi Z, Yalan L, Renlong J, Kang L, Wei L, Houguang L, Wen L, Yuehua Q, Xuanyi L. cAMP-Epac1 signaling is activated in DDAVP-induced endolymphatic hydrops of guinea pigs. Braz J Otorhinolaryngol 2023; 89:469-476. [PMID: 37116375 PMCID: PMC10165185 DOI: 10.1016/j.bjorl.2023.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 02/07/2023] [Accepted: 03/03/2023] [Indexed: 03/16/2023] Open
Abstract
OBJECTIVE To explore whether Cyclic Adenosine Monophosphate (cAMP)-Epac1 signaling is activated in 1-Desamino-8-D-arginine-Vasopressin-induced Endolymphatic Hydrops (DDAVP-induced EH) and to provide new insight for further in-depth study of DDAVP-induced EH. METHODS Eighteen healthy, red-eyed guinea pigs (36 ears) weighing 200-350 g were randomly divided into three groups: the control group, which received intraperitoneal injection of sterile saline (same volume as that in the other two groups) for 7 consecutive days; the DDAVP-7d group, which received intraperitoneal injection of 10 mg/mL/kg DDAVP for 7 consecutive days; and the DDAVP-14d group, which received intraperitoneal injection of 10 μg/mL/kg DDAVP for 14 consecutive days. After successful modeling, all animals were sacrificed, and cochlea tissues were collected to detect the mRNA and protein expression of the exchange protein directly activated by cAMP-1 and 2 (Epac1, Epac2), and Repressor Activator Protein-1 (Rap1) by Reverse Transcription (RT)-PCR and western blotting, respectively. RESULTS Compared to the control group, the relative mRNA expression of Epac1, Epac2, Rap1A, and Rap1B in the cochlea tissue of the DDAVP-7d group was significantly higher (p < 0.05), while no significant difference in Rap1 GTPase activating protein (Rap1gap) mRNA expression was found between the two groups. The relative mRNA expression of Epac1, Rap1A, Rap1B, and Rap1gap in the cochlea tissue of the DDAVP-14d group was significantly higher than that of the control group (p < 0.05), while no significant difference in Epac2 mRNA expression was found between the DDAVP-14d and control groups. Comparison between the DDAVP-14d and DDAVP-7d groups showed that the DDAVP-14d group had significantly lower Epac2 and Rap1A (p < 0.05) and higher Rap1gap (p < 0.05) mRNA expression in the cochlea tissue than that of the DDAVP-7d group, while no significant differences in Epac1 and Rap1B mRNA expression were found between the two groups. Western blotting showed that Epac1 protein expression in the cochlea tissue was the highest in the DDAVP-14d group, followed by that in the DDAVP-7d group, and was the lowest in the control group, showing significant differences between groups (p < 0.05); Rap1 protein expression in the cochlea tissue was the highest in the DDAVP-7d group, followed by the DDAVP-14d group, and was the lowest in the control group, showing significant differences between groups (p < 0.05); no significant differences in Epac2 protein expression in the cochlea tissue were found among the three groups. CONCLUSION DDAVP upregulated Epac1 protein expression in the guinea pig cochlea, leading to activation of the inner ear cAMP-Epac1 signaling pathway. This may be an important mechanism by which DDAVP regulates endolymphatic metabolism to induce EH and affect inner ear function. OXFORD CENTRE FOR EVIDENCE-BASED MEDICINE 2011 LEVELS OF EVIDENCE: Level 5.
Collapse
Affiliation(s)
- Wang Chuan
- The Suqian Clinical College of Xuzhou Medical University, Department of Otorhinolaryngology-Head and Neck Surgery, Suqian, China; Affiliated Hospital of Xuzhou Medical University, Department of Otorhinolaryngology-Head and Neck Surgery, Xuzhou, China; Xuzhou Medical University, Institute of Audiology and Balance Science, Xuzhou, China; Xuzhou Medical University, Artificial Auditory Laboratory of Jiangsu Province, Xuzhou, China
| | - Li Yuan
- Affiliated Hospital of Xuzhou Medical University, Department of Radiology, Xuzhou, China
| | - Jiang Wen
- Affiliated Hospital of Xuzhou Medical University, Department of Otorhinolaryngology-Head and Neck Surgery, Xuzhou, China; Xuzhou Medical University, Institute of Audiology and Balance Science, Xuzhou, China; Xuzhou Medical University, Artificial Auditory Laboratory of Jiangsu Province, Xuzhou, China
| | - Zeng Jianwei
- Affiliated Hospital of Xuzhou Medical University, Department of Radiology, Xuzhou, China
| | - Wang Caiji
- Affiliated Hospital of Xuzhou Medical University, Department of Otorhinolaryngology-Head and Neck Surgery, Xuzhou, China; Xuzhou Medical University, Institute of Audiology and Balance Science, Xuzhou, China; Xuzhou Medical University, Artificial Auditory Laboratory of Jiangsu Province, Xuzhou, China
| | - Zhao Zeqi
- Affiliated Hospital of Xuzhou Medical University, Department of Otorhinolaryngology-Head and Neck Surgery, Xuzhou, China; Xuzhou Medical University, Institute of Audiology and Balance Science, Xuzhou, China; Xuzhou Medical University, Artificial Auditory Laboratory of Jiangsu Province, Xuzhou, China
| | - Li Yalan
- Gulou Hospital Affiliated to Medical College of Nanjing University, Department of Otolaryngology Head and Neck Surgery, Nanjing, China
| | - Ji Renlong
- Affiliated Hospital of Xuzhou Medical University, Department of Otorhinolaryngology-Head and Neck Surgery, Xuzhou, China; Xuzhou Medical University, Institute of Audiology and Balance Science, Xuzhou, China; Xuzhou Medical University, Artificial Auditory Laboratory of Jiangsu Province, Xuzhou, China
| | - Li Kang
- Affiliated Hospital of Xuzhou Medical University, Department of Otorhinolaryngology-Head and Neck Surgery, Xuzhou, China; Xuzhou Medical University, Institute of Audiology and Balance Science, Xuzhou, China; Xuzhou Medical University, Artificial Auditory Laboratory of Jiangsu Province, Xuzhou, China
| | - Li Wei
- Fudan University, Hearing Research Key Lab of Health Ministry of China, Eye and Ear Nose and Throat Hospital, Department of Otology and Skull Base Surgery, Shanghai, China
| | - Liu Houguang
- China University of Mining and Technology, School of Mechatronic Engineering, Xuzhou, China
| | - Liu Wen
- Xuzhou Medical University, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou, China; Affiliated Hospital of Xuzhou Medical University, Department of Otorhinolaryngology-Head and Neck Surgery, Xuzhou, China
| | - Qiao Yuehua
- Affiliated Hospital of Xuzhou Medical University, Department of Otorhinolaryngology-Head and Neck Surgery, Xuzhou, China; Xuzhou Medical University, Institute of Audiology and Balance Science, Xuzhou, China; Xuzhou Medical University, Artificial Auditory Laboratory of Jiangsu Province, Xuzhou, China
| | - Li Xuanyi
- Affiliated Hospital of Xuzhou Medical University, Department of Otorhinolaryngology-Head and Neck Surgery, Xuzhou, China; Xuzhou Medical University, Institute of Audiology and Balance Science, Xuzhou, China; Xuzhou Medical University, Artificial Auditory Laboratory of Jiangsu Province, Xuzhou, China.
| |
Collapse
|
26
|
Targeting Human Proteins for Antiviral Drug Discovery and Repurposing Efforts: A Focus on Protein Kinases. Viruses 2023; 15:v15020568. [PMID: 36851782 PMCID: PMC9966946 DOI: 10.3390/v15020568] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 02/22/2023] Open
Abstract
Despite the great technological and medical advances in fighting viral diseases, new therapies for most of them are still lacking, and existing antivirals suffer from major limitations regarding drug resistance and a limited spectrum of activity. In fact, most approved antivirals are directly acting antiviral (DAA) drugs, which interfere with viral proteins and confer great selectivity towards their viral targets but suffer from resistance and limited spectrum. Nowadays, host-targeted antivirals (HTAs) are on the rise, in the drug discovery and development pipelines, in academia and in the pharmaceutical industry. These drugs target host proteins involved in the virus life cycle and are considered promising alternatives to DAAs due to their broader spectrum and lower potential for resistance. Herein, we discuss an important class of HTAs that modulate signal transduction pathways by targeting host kinases. Kinases are considered key enzymes that control virus-host interactions. We also provide a synopsis of the antiviral drug discovery and development pipeline detailing antiviral kinase targets, drug types, therapeutic classes for repurposed drugs, and top developing organizations. Furthermore, we detail the drug design and repurposing considerations, as well as the limitations and challenges, for kinase-targeted antivirals, including the choice of the binding sites, physicochemical properties, and drug combinations.
Collapse
|
27
|
Yoshie M, Ohishi K, Ishikawa G, Tsuru A, Kusama K, Azumi M, Tamura K. Small GTP-binding protein Rap1 mediates EGF and HB-EGF signaling and modulates EGF receptor expression in HTR-8/SVneo extravillous trophoblast cells. Reprod Med Biol 2023; 22:e12537. [PMID: 37614815 PMCID: PMC10442520 DOI: 10.1002/rmb2.12537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 08/06/2023] [Accepted: 08/08/2023] [Indexed: 08/25/2023] Open
Abstract
Purpose Extravillous trophoblasts (EVTs) invade the endometrium to establish a fetomaternal interaction during pregnancy. Epidermal growth factor (EGF) and heparin-binding EGF-like growth factor (HB-EGF) stimulate EVT invasion by binding to the EGF receptor (EGFR). We examined the role of the small GTP-binding protein Rap1 in EGF- and HB-EGF-stimulated EVT invasion. Methods Expression of Rap1 in the first-trimester placenta was examined by immunohistochemistry. Effect of EGF or HB-EGF on Rap1 activation (GTP-Rap1) and Rap1 knockdown on invasion was assessed in EVT cell line (HTR-8/SVneo). In addition, effect of Rap1 knockdown and Rap1GAP (a Rap1 inactivator) overexpression on the activation of EGF signaling and EGFR expression were examined. Results Rap1 was expressed by EVTs, villous cytotrophoblasts, and syncytiotrophoblasts in the placenta. EGF and HB-EGF activated Rap1 and promoted invasion of HTR-8/SVneo, and these effects were inhibited by Rap1 knockdown. The EGF- and HB-EGF-induced phosphorylation of AKT, ERK1/2, p38MAPK, and Src was inhibited by Rap1 knockdown. Furthermore, the knockdown of Rap1 reduced the EGFR protein level. Overexpression of Rap1GAP repressed EGF- and HB-EGF-induced Rap1 activation and reduced EGFR expression. Conclusion Rap1 may function as a mediator of EGF and HB-EGF signaling pathways and can modulate EGFR expression in EVTs during placental development.
Collapse
Affiliation(s)
- Mikihiro Yoshie
- Department of Endocrine PharmacologyTokyo University of Pharmacy and Life SciencesTokyoJapan
| | - Kensuke Ohishi
- Department of Endocrine PharmacologyTokyo University of Pharmacy and Life SciencesTokyoJapan
| | - Gen Ishikawa
- Department of ObstetricsMiyagi Children's HospitalSendaiJapan
| | - Atsuya Tsuru
- Department of Endocrine PharmacologyTokyo University of Pharmacy and Life SciencesTokyoJapan
| | - Kazuya Kusama
- Department of Endocrine PharmacologyTokyo University of Pharmacy and Life SciencesTokyoJapan
| | - Mana Azumi
- Department of Endocrine PharmacologyTokyo University of Pharmacy and Life SciencesTokyoJapan
| | - Kazuhiro Tamura
- Department of Endocrine PharmacologyTokyo University of Pharmacy and Life SciencesTokyoJapan
| |
Collapse
|
28
|
Shang D, Lan T, Wang Y, Li X, Liu Q, Dong H, Xu B, Cheng H, Zhou R. PGCLCs of human 45,XO reveal pathogenetic pathways of neurocognitive and psychosocial disorders. Cell Biosci 2022; 12:194. [DOI: 10.1186/s13578-022-00925-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/03/2022] [Indexed: 12/03/2022] Open
Abstract
Abstract
Background
Neurocognitive disorders and psychosocial difficulties are common in patients with Turner syndrome and multiple neurodegenerative diseases, yet there is no effective cure. Human primordial germ cells (hPGCs) are pluripotent germline stem cells in early embryo, which pass genetic information from one generation to the next, whereas all somatic cells will die along with the end of life. However, it is not known whether patient hPGCs with Turner syndrome contain information of neurocognitive and psychosocial illness.
Results
In this report, we used a high-density of culture system of embryoids derived from iPSCs of a patient with Turner syndrome to ask how pathogenetic pathways are associated with onset of neurocognitive and psychosocial disorders. The hPGC-Like Cells (hPGCLCs) were in vitro specified from iPSCs of 45,XO, 46,XX and 46,XY by the high-density induction of embryoids. Amazingly, we found that the specification process of the hPGCLCs in 45,XO, compared to those in 46,XX and 46,XY, enriched several common pathogenetic pathways regulating neurocognitive and psychosocial disorders, that shared among multiple neurodegenerative diseases and Turner syndrome. The downregulated chemical synaptic transmission pathways, including glutamatergic, GABAergic, and nicotine cholinergic synapses, indicated synaptic dysfunctions, while upregulated pathways that were associated with imbalance of mitochondrial respiratory chain complexes and apoptosis, may contribute to neuronal dysfunctions. Notably, downregulation of three types of ubiquitin ligases E1-E2-E3 and lysosome-associated sulfatases and RAB9A, owing to haploinsufficiency and parental preference of the X chromosome expression, indicated that two pathways of cellular degradation, lysosome and ubiquitin–proteasome, were impaired in the specification process of 45,XO hPGCLCs. This would lead to accumulation of undesired proteins and aggregates, which is a typically pathological hallmark in neurodegenerative diseases.
Conclusions
Our data suggest that the specification process of the hPGCLCs in 45,XO, compared to those in 46,XX and 46,XY, enriched pathogenetic pathways that are associated with the onset of neurocognitive and psychosocial disorders.
Collapse
|
29
|
Wang X, Wang J, Raza SHA, Deng J, Ma J, Qu X, Yu S, Zhang D, Alshammari AM, Almohaimeed HM, Zan L. Identification of the hub genes related to adipose tissue metabolism of bovine. Front Vet Sci 2022; 9:1014286. [PMID: 36439361 PMCID: PMC9682410 DOI: 10.3389/fvets.2022.1014286] [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: 08/08/2022] [Accepted: 09/16/2022] [Indexed: 11/11/2022] Open
Abstract
Due to the demand for high-quality animal protein, there has been consistent interest in how to obtain more high-quality beef. As well-known, the adipose content of beef has a close connection with the taste and quality of beef, and cattle with different energy or protein diet have corresponding effects on the lipid metabolism of beef. Thus, we performed weighted gene co-expression network analysis (WGCNA) with subcutaneous adipose genes from Norwegian red heifers fed different diets to identify hub genes regulating bovine lipid metabolism. For this purpose, the RNA sequencing data of subcutaneous adipose tissue of 12-month-old Norwegian red heifers (n = 48) with different energy or protein levels were selected from the GEO database, and 7,630 genes with the largest variation were selected for WGCNA analysis. Then, three modules were selected as hub genes candidate modules according to the correlation between modules and phenotypes, including pink, magenta and grey60 modules. GO and KEGG enrichment analysis showed that genes were related to metabolism, and participated in Rap, MAPK, AMPK, VEGF signaling pathways, and so forth. Combined gene interaction network analysis using Cytoscape software, eight hub genes of lipid metabolism were identified, including TIA1, LOC516108, SNAPC4, CPSF2, ZNF574, CLASRP, MED15 and U2AF2. Further, the expression levels of hub genes in the cattle tissue were also measured to verify the results, and we found hub genes in higher expression in muscle and adipose tissue in adult cattle. In summary, we predicted the key genes of lipid metabolism in the subcutaneous adipose tissue that were affected by the intake of various energy diets to find the hub genes that coordinate lipid metabolism, which provide a theoretical basis for regulating beef quality.
Collapse
Affiliation(s)
- Xiaohui Wang
- College of Animal Science and Technology, Northwest A&F University, Xianyang, China
| | - Jianfang Wang
- College of Animal Science and Technology, Northwest A&F University, Xianyang, China
| | | | - Jiahan Deng
- College of Animal Science and Technology, Northwest A&F University, Xianyang, China
| | - Jing Ma
- College of Animal Science and Technology, Northwest A&F University, Xianyang, China
| | - Xiaopeng Qu
- College of Animal Science and Technology, Northwest A&F University, Xianyang, China
| | - Shengchen Yu
- College of Animal Science and Technology, Northwest A&F University, Xianyang, China
| | - Dianqi Zhang
- College of Animal Science and Technology, Northwest A&F University, Xianyang, China
| | | | - Hailah M. Almohaimeed
- Department of Basic Science, College of Medicine, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Linsen Zan
- College of Animal Science and Technology, Northwest A&F University, Xianyang, China
- National Beef Cattle Improvement Center, Northwest A&F University, Xianyang, China
- *Correspondence: Linsen Zan
| |
Collapse
|
30
|
Yadav V, Sharma K, Bhattacharya S, Talwar P, Purohit PK, Saini N. RETRACTED: hsa-miR-23a~27a~24-2 cluster members inhibit aggressiveness of breast cancer cells by commonly targeting NCOA1, NLK and RAP1B. Life Sci 2022; 307:120906. [PMID: 36007610 DOI: 10.1016/j.lfs.2022.120906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 08/10/2022] [Accepted: 08/18/2022] [Indexed: 11/28/2022]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editor-in-Chief. The corresponding author notified the journal of three examples of image duplication within the published article (two in Figure 3D and one in Figure 4A), and requested a corrigendum. As per journal policy when considering corrigendum requests, the journal requested the authors to provide source data relating to these affected figures. The editorial team noticed 12 additional suspected image duplications within the supplied source data and the corresponding author was informed. Upon submission of revised source data, the editorial team noticed two new suspected image duplications. The editorial team have concerns about the provenance of the data and therefore the Editor-in-Chief decided to retract the article.
Collapse
Affiliation(s)
- Vikas Yadav
- Functional Genomics Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), Mall Road, 110007, Delhi, India
| | - Kritika Sharma
- Functional Genomics Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), Mall Road, 110007, Delhi, India
| | - Sushant Bhattacharya
- Functional Genomics Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), Mall Road, 110007, Delhi, India
| | - Puneet Talwar
- Institute of Human Behaviour & Allied Sciences (IHBAS), Delhi, India
| | - Paresh Kumar Purohit
- Functional Genomics Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), Mall Road, 110007, Delhi, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Neeru Saini
- Functional Genomics Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), Mall Road, 110007, Delhi, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
31
|
Miller D, Saeed A, Nelson AC, Bower M, Aggarwal A. Third reported patient with RAP1B-related syndromic thrombocytopenia and novel clinical findings. Am J Med Genet A 2022; 188:2808-2814. [PMID: 35451551 PMCID: PMC9543813 DOI: 10.1002/ajmg.a.62760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/28/2022] [Accepted: 04/01/2022] [Indexed: 01/25/2023]
Abstract
RAP1B is a RAS-superfamily small GTP-binding protein involved in numerous cell processes. Pathogenic gain-of-function variants in this gene have been associated with RAP1B-related syndromic thrombocytopenia, an ultrarare disorder characterized by hematologic abnormalities, neurodevelopmental delays, growth delay, and congenital birth defects including cardiovascular, genitourinary, neurologic, and skeletal systems. We report a 23-year-old male with a novel, de novo RAP1B gain-of-function variant identified on genome sequencing. This is the third reported case which expands the molecular and phenotypic spectrum of RAP1B-related syndromic thrombocytopenia.
Collapse
Affiliation(s)
| | - Azhar Saeed
- Department of Laboratory Medicine and PathologyUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Andrew C. Nelson
- Department of Laboratory Medicine and PathologyUniversity of MinnesotaMinneapolisMinnesotaUSA
| | | | - Anjali Aggarwal
- Division of Genetics and Metabolism, Department of PediatricsUniversity of MinnesotaMinneapolisMinnesotaUSA
| |
Collapse
|
32
|
Life at the periphery: what makes CHO cells survival talents. Appl Microbiol Biotechnol 2022; 106:6157-6167. [PMID: 36038753 PMCID: PMC9468092 DOI: 10.1007/s00253-022-12123-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 08/02/2022] [Accepted: 08/03/2022] [Indexed: 11/24/2022]
Abstract
Abstract
The production of biopharmaceuticals relies on robust cell systems that can produce recombinant proteins at high levels and grow and survive in the stressful bioprocess environment. Chinese hamster ovary cells (CHO) as the main production hosts offer a variety of advantages including robust growth and survival in a bioprocess environment. Cell surface proteins are of special interest for the understanding of how CHO cells react to their environment while maintaining growth and survival phenotypes, since they enable cellular reactions to external stimuli and potentially initiate signaling pathways. To provide deeper insight into functions of this special cell surface sub-proteome, pathway enrichment analysis of the determined CHO surfaceome was conducted. Enrichment of growth/ survival-pathways such as the phosphoinositide-3-kinase (PI3K)–protein kinase B (AKT), mitogen-activated protein kinase (MAPK), Janus kinase/signal transducers and activators of transcription (JAK-STAT), and RAP1 pathways were observed, offering novel insights into how cell surface receptors and ligand-mediated signaling enable the cells to grow and survive in a bioprocess environment. When supplementing surfaceome data with RNA expression data, several growth/survival receptors were shown to be co-expressed with their respective ligands and thus suggesting self-induction mechanisms, while other receptors or ligands were not detectable. As data about the presence of surface receptors and their associated expressed ligands may serve as base for future studies, further pathway characterization will enable the implementation of optimization strategies to further enhance cellular growth and survival behavior. Key points • PI3K/AKT, MAPK, JAK-STAT, and RAP1 pathway receptors are enriched on the CHO cell surface and downstream pathways present on mRNA level. • Detected pathways indicate strong CHO survival and growth phenotypes. • Potential self-induction of surface receptors and respective ligands. Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1007/s00253-022-12123-6.
Collapse
|
33
|
Wang Y, Spolitu S, Zadroga JA, Sarecha AK, Ozcan L. Hepatocyte Rap1a contributes to obesity- and statin-associated hyperglycemia. Cell Rep 2022; 40:111259. [PMID: 36001955 PMCID: PMC9446800 DOI: 10.1016/j.celrep.2022.111259] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 05/26/2022] [Accepted: 08/03/2022] [Indexed: 12/28/2022] Open
Abstract
Excessive hepatic glucose production contributes to the development of hyperglycemia and is a key feature of type 2 diabetes. Here, we report that activation of hepatocyte Rap1a suppresses gluconeogenic gene expression and glucose production, whereas Rap1a silencing stimulates them. Rap1a activation is suppressed in obese mouse liver, and restoring its activity improves glucose intolerance. As Rap1a′s membrane localization and activation depends on its geranylgeranylation, which is inhibited by statins, we show that statin-treated hepatocytes and the human liver have lower active-Rap1a levels. Similar to Rap1a inhibition, statins stimulate hepatic gluconeogenesis and increase fasting blood glucose in obese mice. Geranylgeraniol treatment, which acts as the precursor for geranylgeranyl isoprenoids, restores Rap1a activity and improves statin-mediated glucose intolerance. Mechanistically, Rap1a activation induces actin polymerization, which suppresses gluconeogenesis by Akt-mediated FoxO1 inhibition. Thus, Rap1a regulates hepatic glucose homeostasis, and blocking its activity, via lowering geranylgeranyl isoprenoids, contributes to statin-induced glucose intolerance. Wang et al. show that activation of hepatic Rap1a suppresses gluconeogenic gene expression and improves glucose intolerance via Akt-mediated FoxO1 inhibition. Statins lower intracellular isoprenoid levels and inhibit Rap1a activation, which contributes to their hyperglycemic effect. These findings identify a role of hepatic Rap1a in obesity- and statin-associated glucose homeostasis.
Collapse
Affiliation(s)
- Yating Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Stefano Spolitu
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - John A Zadroga
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Amesh K Sarecha
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Lale Ozcan
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
34
|
Gao N, Raduka A, Rezaee F. Respiratory syncytial virus disrupts the airway epithelial barrier by decreasing cortactin and destabilizing F-actin. J Cell Sci 2022; 135:jcs259871. [PMID: 35848790 PMCID: PMC9481929 DOI: 10.1242/jcs.259871] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 07/11/2022] [Indexed: 01/26/2023] Open
Abstract
Respiratory syncytial virus (RSV) infection is the leading cause of acute lower respiratory tract infection in young children worldwide. Our group recently revealed that RSV infection disrupts the airway epithelial barrier in vitro and in vivo. However, the underlying molecular pathways were still elusive. Here, we report the critical roles of the filamentous actin (F-actin) network and actin-binding protein cortactin in RSV infection. We found that RSV infection causes F-actin depolymerization in 16HBE cells, and that stabilizing the F-actin network in infected cells reverses the epithelial barrier disruption. RSV infection also leads to significantly decreased cortactin in vitro and in vivo. Cortactin-knockout 16HBE cells presented barrier dysfunction, whereas overexpression of cortactin protected the epithelial barrier against RSV. The activity of Rap1 (which has Rap1A and Rap1B forms), one downstream target of cortactin, declined after RSV infection as well as in cortactin-knockout cells. Moreover, activating Rap1 attenuated RSV-induced epithelial barrier disruption. Our study proposes a key mechanism in which RSV disrupts the airway epithelial barrier via attenuating cortactin expression and destabilizing the F-actin network. The identified pathways will provide new targets for therapeutic intervention toward RSV-related disease. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Nannan Gao
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA
| | - Andjela Raduka
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA
| | - Fariba Rezaee
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA
- Center for Pediatric Pulmonary Medicine, Cleveland Clinic Children's, Cleveland, Ohio 44195, USA
| |
Collapse
|
35
|
Role of an FNIP Repeat Domain-Containing Protein Encoded by Megavirus Baoshan during Viral Infection. J Virol 2022; 96:e0081322. [PMID: 35762756 PMCID: PMC9327691 DOI: 10.1128/jvi.00813-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
FNIP repeat domain-containing protein (FNIP protein) is a little-studied atypical leucine-rich repeat domain-containing protein found in social amoebae and mimiviruses. Here, a recently reported mimivirus of lineage C, Megavirus baoshan, was analyzed for FNIP protein genes. A total of 82 FNIP protein genes were identified, each containing up to 26 copies of the FNIP repeat, and mostly having an F-box domain at the N terminus. Both nucleotide and amino acid sequences of FNIP repeat were highly conserved. Most of the FNIP protein genes clustered together tandemly in groups of two to 14 genes. Nearly all FNIP protein genes shared similar expression patterns and were expressed 4 to 9 h postinfection. A typical viral FNIP protein, Mb0983, was selected for functional analysis. Protein interactome analysis identified two small GTPases, Rap1B and Rab7A, that interacted with Mb0983 in cytoplasm. The overexpression of Mb0983 in Acanthamoeba castellanii accelerated the degradation of Rap1B and Rab7A during viral infection. Mb0983 also interacted with host SKP1 and cullin-1, which were conserved components of the SKP1-cullin-1-F-box protein (SCF)-type ubiquitin E3 ligase complex. Deletion of the F-box domain of Mb0983 not only abolished its interaction with SKP1 and cullin-1 but also returned the speed of Rap1B and Rab7A degradation to normal in infected A. castellanii. These results suggested that Mb0983 is a part of the SCF-type ubiquitin E3 ligase complex and plays a role in the degradation of Rap1B and Rab7A. They also implied that other viral F-box-containing FNIP proteins might have similar effects on various host proteins. IMPORTANCE Megavirus baoshan encodes 82 FNIP proteins, more than any other reported mimiviruses. Their genetic and transcriptional features suggest that they are important for virus infection and adaption. Since most mimiviral FNIP proteins have the F-box domain, they were predicted to be involved in protein ubiquitylation. FNIP protein Mb0983 interacted with host SKP1 and cullin-1 through the F-box domain, supporting the idea that it is a part of the SCF-type ubiquitin E3 ligase complex. The substrates of Mb0983 for degradation were identified as the host small GTPases Rap1B and Rab7A. Combining the facts of the presence of a large number of FNIP genes in megavirus genomes, the extremely high expression level of the viral ubiquitin gene, and the reported observation that 35% of megavirus-infected amoeba cells died without productive infection, it is likely that megavirus actively explores the host ubiquitin-proteasome pathway in infection and that viral FNIP proteins play roles in the process.
Collapse
|
36
|
Integrin Regulators in Neutrophils. Cells 2022; 11:cells11132025. [PMID: 35805108 PMCID: PMC9266208 DOI: 10.3390/cells11132025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/17/2022] [Accepted: 06/22/2022] [Indexed: 02/01/2023] Open
Abstract
Neutrophils are the most abundant leukocytes in humans and are critical for innate immunity and inflammation. Integrins are critical for neutrophil functions, especially for their recruitment to sites of inflammation or infections. Integrin conformational changes during activation have been heavily investigated but are still not fully understood. Many regulators, such as talin, Rap1-interacting adaptor molecule (RIAM), Rap1, and kindlin, are critical for integrin activation and might be potential targets for integrin-regulating drugs in treating inflammatory diseases. In this review, we outline integrin activation regulators in neutrophils with a focus on the above critical regulators, as well as newly discovered modulators that are involved in integrin activation.
Collapse
|
37
|
Wang A, Li Z, Zhuo S, Gao F, Zhang H, Zhang Z, Ren G, Ma X. Mechanisms of Cardiorenal Protection With SGLT2 Inhibitors in Patients With T2DM Based on Network Pharmacology. Front Cardiovasc Med 2022; 9:857952. [PMID: 35677689 PMCID: PMC9169967 DOI: 10.3389/fcvm.2022.857952] [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: 01/24/2022] [Accepted: 05/04/2022] [Indexed: 11/24/2022] Open
Abstract
Purpose Sodium-glucose cotransporter 2 (SGLT2) inhibitors have cardiorenal protective effects regardless of whether they are combined with type 2 diabetes mellitus, but their specific pharmacological mechanisms remain undetermined. Materials and Methods We used databases to obtain information on the disease targets of “Chronic Kidney Disease,” “Heart Failure,” and “Type 2 Diabetes Mellitus” as well as the targets of SGLT2 inhibitors. After screening the common targets, we used Cytoscape 3.8.2 software to construct SGLT2 inhibitors' regulatory network and protein-protein interaction network. The clusterProfiler R package was used to perform gene ontology functional analysis and Kyoto encyclopedia of genes and genomes pathway enrichment analyses on the target genes. Molecular docking was utilized to verify the relationship between SGLT2 inhibitors and core targets. Results Seven different SGLT2 inhibitors were found to have cardiorenal protective effects on 146 targets. The main mechanisms of action may be associated with lipid and atherosclerosis, MAPK signaling pathway, Rap1 signaling pathway, endocrine resistance, fluid shear stress, atherosclerosis, TNF signaling pathway, relaxin signaling pathway, neurotrophin signaling pathway, and AGEs-RAGE signaling pathway in diabetic complications were related. Docking of SGLT2 inhibitors with key targets such as GAPDH, MAPK3, MMP9, MAPK1, and NRAS revealed that these compounds bind to proteins spontaneously. Conclusion Based on pharmacological networks, this study elucidates the potential mechanisms of action of SGLT2 inhibitors from a systemic and holistic perspective. These key targets and pathways will provide new ideas for future studies on the pharmacological mechanisms of cardiorenal protection by SGLT2 inhibitors.
Collapse
Affiliation(s)
- Anzhu Wang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhendong Li
- Qingdao West Coast New Area People's Hospital, Qingdao, China
| | - Sun Zhuo
- Qingdao West Coast New Area People's Hospital, Qingdao, China
| | - Feng Gao
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hongwei Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhibo Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Gaocan Ren
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaochang Ma
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
- *Correspondence: Xiaochang Ma
| |
Collapse
|
38
|
Parkes MD, Halloran K, Hirji A, Pon S, Weinkauf J, Timofte IL, Snell GI, Westall GP, Havlin J, Lischke R, Zajacová A, Hachem R, Kreisel D, Levine D, Kubisa B, Piotrowska M, Juvet S, Keshavjee S, Jaksch P, Klepetko W, Halloran PF. Transcripts associated with chronic lung allograft dysfunction in transbronchial biopsies of lung transplants. Am J Transplant 2022; 22:1054-1072. [PMID: 34850543 DOI: 10.1111/ajt.16895] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 10/14/2021] [Accepted: 11/07/2021] [Indexed: 01/25/2023]
Abstract
Transplanted lungs suffer worse outcomes than other organ transplants with many developing chronic lung allograft dysfunction (CLAD), diagnosed by physiologic changes. Histology of transbronchial biopsies (TBB) yields little insight, and the molecular basis of CLAD is not defined. We hypothesized that gene expression in TBBs would reveal the nature of CLAD and distinguish CLAD from changes due simply to time posttransplant. Whole-genome mRNA profiling was performed with microarrays in 498 prospectively collected TBBs from the INTERLUNG study, 90 diagnosed as CLAD. Time was associated with increased expression of inflammation genes, for example, CD1E and immunoglobulins. After correcting for time, CLAD manifested not as inflammation but as parenchymal response-to-wounding, with increased expression of genes such as HIF1A, SERPINE2, and IGF1 that are increased in many injury and disease states and cancers, associated with development, angiogenesis, and epithelial response-to-wounding in pathway analysis. Fibrillar collagen genes were increased in CLAD, indicating matrix changes, and normal transcripts were decreased-dedifferentiation. Gene-based classifiers predicted CLAD with AUC 0.70 (no time-correction) and 0.87 (time-corrected). CLAD related gene sets and classifiers were strongly prognostic for graft failure and correlated with CLAD stage. Thus, in TBBs, molecular changes indicate that CLAD primarily reflects severe parenchymal injury-induced changes and dedifferentiation.
Collapse
Affiliation(s)
| | | | - Alim Hirji
- University of Alberta, Edmonton, Alberta, Canada
| | - Shane Pon
- University of Alberta, Edmonton, Alberta, Canada
| | | | | | - Greg I Snell
- Alfred Hospital Lung Transplant Service, Melbourne, Australia
| | - Glen P Westall
- Alfred Hospital Lung Transplant Service, Melbourne, Australia
| | - Jan Havlin
- University Hospital Motol, Prague, Czech Republic
| | | | | | - Ramsey Hachem
- Washington University in St Louis, St. Louis, Missouri, USA
| | - Daniel Kreisel
- Washington University in St Louis, St. Louis, Missouri, USA
| | - Deborah Levine
- University of Texas San Antonio, San Antonio, Texas, USA
| | - Bartosz Kubisa
- Pomeranian Medical University of Szczecin, Szczecin, Poland
| | | | - Stephen Juvet
- Toronto Lung Transplant Program, University Health Network, Toronto, Ontario, Canada
| | - Shaf Keshavjee
- Toronto Lung Transplant Program, University Health Network, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
39
|
Maywald ML, Picciotto C, Lepa C, Bertgen L, Yousaf FS, Ricker A, Klingauf J, Krahn MP, Pavenstädt H, George B. Rap1 Activity Is Essential for Focal Adhesion and Slit Diaphragm Integrity. Front Cell Dev Biol 2022; 10:790365. [PMID: 35372328 PMCID: PMC8972170 DOI: 10.3389/fcell.2022.790365] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 02/24/2022] [Indexed: 11/24/2022] Open
Abstract
Glomerular podocytes build, with their intercellular junctions, part of the kidney filter. The podocyte cell adhesion protein, nephrin, is essential for developing and maintaining slit diaphragms as functional loss in humans results in heavy proteinuria. Nephrin expression and function are also altered in many adult-onset glomerulopathies. Nephrin signals from the slit diaphragm to the actin cytoskeleton and integrin β1 at focal adhesions by recruiting Crk family proteins, which can interact with the Rap guanine nucleotide exchange factor 1 C3G. As Rap1 activity affects focal adhesion formation, we hypothesize that nephrin signals via Rap1 to integrin β. To address this issue, we combined Drosophila in vivo and mammalian cell culture experiments. We find that Rap1 is necessary for correct targeting of integrin β to focal adhesions in Drosophila nephrocytes, which also form slit diaphragm-like structures. In the fly, the Rap1 activity is important for signaling of the nephrin ortholog to integrin β, as well as for nephrin-dependent slit diaphragm integrity. We show by genetic interaction experiments that Rap1 functions downstream of nephrin signaling to integrin β and downstream of nephrin signaling necessary for slit diaphragm integrity. Similarly, in human podocyte culture, nephrin activation results in increased activation of Rap1. Thus, Rap1 is necessary for downstream signal transduction of nephrin to integrin β.
Collapse
Affiliation(s)
- Mee-Ling Maywald
- Medizinische Klinik D, University Hospital Münster, Münster, Germany
| | - Cara Picciotto
- Medizinische Klinik D, University Hospital Münster, Münster, Germany
| | - Carolin Lepa
- Medizinische Klinik D, University Hospital Münster, Münster, Germany
| | - Luisa Bertgen
- Medizinische Klinik D, University Hospital Münster, Münster, Germany
| | | | - Andrea Ricker
- Institute of Medical Physics and Biophysics, Westfälische Wilhelms-University Münster, Münster, Germany
| | - Jürgen Klingauf
- Institute of Medical Physics and Biophysics, Westfälische Wilhelms-University Münster, Münster, Germany
| | - Michael P. Krahn
- Medizinische Klinik D, Medical Cell Biology, University Hospital Münster, Münster, Germany
| | | | - Britta George
- Medizinische Klinik D, University Hospital Münster, Münster, Germany
- *Correspondence: Britta George,
| |
Collapse
|
40
|
Rezaie J, Akbari A, Rahbarghazi R. Inhibition of extracellular vesicle biogenesis in tumor cells: A possible way to reduce tumorigenesis. Cell Biochem Funct 2022; 40:248-262. [PMID: 35285964 DOI: 10.1002/cbf.3695] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/14/2022] [Accepted: 02/21/2022] [Indexed: 12/17/2022]
Abstract
Most eukaryotic cells secrete extracellular vesicles (EVs), which contribute to intracellular communication through transferring different biomolecules such as proteins, RNAs, and lipids to cells. Two main types of EVs are exosomes and microvesicles. Exosomes originate from multivesicular bodies, while microvesicles are shed from the plasma membrane. Mechanisms of exosomes and microvesicle biogenesis/trafficking are complex and many molecules are involved in their biogenesis and secretion. Tumor-derived EVs contain oncogenic molecules that promote tumor growth, metastasis, immune surveillance, angiogenesis, and chemoresistance. A growing body of evidence indicates various compounds can inhibit biogenesis and secretion of EVs from cells and several experiments were conducted to use EVs-inhibitors for understanding the biology of the cells or for understanding the pathology of several diseases like cancer. However, the nontargeting effects of drugs/inhibitors remain a concern. Our current knowledge of EVs biogenesis and their inhibition from tumor cells may provide an avenue for cancer management. In this review, we shed light on exosomes and microvesicles biogenesis, key roles of tumor-derived EVs, and discuss methods used to inhibition of EVs by different inhibitors.
Collapse
Affiliation(s)
- Jafar Rezaie
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Ali Akbari
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
41
|
Liu Y, Chen L, Liu W, Li D, Zeng J, Tang Q, Zhang Y, Luan F, Zeng N. Cepharanthine Suppresses Herpes Simplex Virus Type 1 Replication Through the Downregulation of the PI3K/Akt and p38 MAPK Signaling Pathways. Front Microbiol 2021; 12:795756. [PMID: 34956164 PMCID: PMC8696181 DOI: 10.3389/fmicb.2021.795756] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 11/08/2021] [Indexed: 12/23/2022] Open
Abstract
Cepharanthine (CEP) is a naturally occurring isoquinoline alkaloid extracted from Stephania cepharantha Hayata. Although its underlying molecular mechanism is not fully understood, this compound is reported as a promising antiviral drug. In the present study, we explore the anti-HSV-1 effects and the underlying molecular mechanisms of CEP in vitro. Our results show that CEP could significantly inhibit the formation of plaque and the expression of viral proteins and exhibit a general suppression of replication-associated genes. Whereas HSV-1 infection increases the expressions of phosphoinositide 3-kinase (PI3K), protein kinase B (Akt), and p38 mitogen-activated protein kinase (p38 MAPK) in host cells, CEP was effective indirectly inhibiting phosphorylation levels of the targets in PI3K/Akt and p38 MAPK signaling pathways. Moreover, CEP markedly decreased G0/G1 phase and increased G2/M phase cells and decreased the expression of cyclin-dependent kinase1 (CDK1) and cyclinB1 in a dose-dependent manner. Additionally, CEP increased apoptosis in infected cells, reduced B cell lymphoma-2 (Bcl-2) protein levels, and increased the protein levels of Bcl-associated X protein (Bax), cleaved-caspase3, and nuclear IκB kinaseα (IκBα). Collectively, CEP could arrest the cell cycle in the G2/M phase and induce apoptosis in infected cells by inhibiting the PI3K/Akt and p38 MAPK signaling pathways, hence further reducing HSV-1 infection and subsequent reproduction.
Collapse
Affiliation(s)
- Yao Liu
- State Key Laboratory of South Western Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,School of Laboratory Medicine, Chengdu Medical College, Chengdu, China
| | - Li Chen
- State Key Laboratory of South Western Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Department of Pharmacy, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Wenjun Liu
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| | - Dan Li
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| | - Jiuseng Zeng
- State Key Laboratory of South Western Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiong Tang
- State Key Laboratory of South Western Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuexin Zhang
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| | - Fei Luan
- State Key Laboratory of South Western Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Nan Zeng
- State Key Laboratory of South Western Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
42
|
Li Y, Deng P, Chen C, Ma Q, Pi H, He M, Lu Y, Gao P, Zhou C, He Z, Zhang Y, Yu Z, Zhang L. 1,800 MHz Radiofrequency Electromagnetic Irradiation Impairs Neurite Outgrowth With a Decrease in Rap1-GTP in Primary Mouse Hippocampal Neurons and Neuro2a Cells. Front Public Health 2021; 9:771508. [PMID: 34881219 PMCID: PMC8646047 DOI: 10.3389/fpubh.2021.771508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 10/29/2021] [Indexed: 12/23/2022] Open
Abstract
Background: With the global popularity of communication devices such as mobile phones, there are increasing concerns regarding the effect of radiofrequency electromagnetic radiation (RF-EMR) on the brain, one of the most important organs sensitive to RF-EMR exposure at 1,800 MHz. However, the effects of RF-EMR exposure on neuronal cells are unclear. Neurite outgrowth plays a critical role in brain development, therefore, determining the effects of 1,800 MHz RF-EMR exposure on neurite outgrowth is important for exploring its effects on brain development. Objectives: We aimed to investigate the effects of 1,800 MHz RF-EMR exposure for 48 h on neurite outgrowth in neuronal cells and to explore the associated role of the Rap1 signaling pathway. Material and Methods: Primary hippocampal neurons from C57BL/6 mice and Neuro2a cells were exposed to 1,800 MHz RF-EMR at a specific absorption rate (SAR) value of 4 W/kg for 48 h. CCK-8 assays were used to determine the cell viability after 24, 48, and 72 h of irradiation. Neurite outgrowth of primary hippocampal neurons (DIV 2) and Neuro2a cells was observed with a 20 × optical microscope and recognized by ImageJ software. Rap1a and Rap1b gene expressions were detected by real-time quantitative PCR. Rap1, Rap1a, Rap1b, Rap1GAP, and p-MEK1/2 protein expressions were detected by western blot. Rap1-GTP expression was detected by immunoprecipitation. The role of Rap1-GTP was assessed by transfecting a constitutively active mutant plasmid (Rap1-Gly_Val-GFP) into Neuro2a cells. Results: Exposure to 1,800 MHz RF-EMR for 24, 48, and 72 h at 4 W/kg did not influence cell viability. The neurite length, primary and secondary neurite numbers, and branch points of primary mouse hippocampal neurons were significantly impaired by 48-h RF-EMR exposure. The neurite-bearing cell percentage and neurite length of Neuro2a cells were also inhibited by 48-h RF-EMR exposure. Rap1 activity was inhibited by 48-h RF-EMR with no detectable alteration in either gene or protein expression of Rap1. The protein expression of Rap1GAP increased after 48-h RF-EMR exposure, while the expression of p-MEK1/2 protein decreased. Overexpression of constitutively active Rap1 reversed the decrease in Rap1-GTP and the neurite outgrowth impairment in Neuro2a cells induced by 1,800 MHz RF-EMR exposure for 48 h. Conclusion: Rap1 activity and related signaling pathways are involved in the disturbance of neurite outgrowth induced by 48-h 1,800 MHz RF-EMR exposure. The effects of RF-EMR exposure on neuronal development in infants and children deserve greater focus.
Collapse
Affiliation(s)
- Yanqi Li
- Key Laboratory of Medical Protection for Electromagnetic Radiation, Department of Occupational Health, Ministry of Education, Third Military Medical University, Chongqing, China
| | - Ping Deng
- Key Laboratory of Medical Protection for Electromagnetic Radiation, Department of Occupational Health, Ministry of Education, Third Military Medical University, Chongqing, China
| | - Chunhai Chen
- Key Laboratory of Medical Protection for Electromagnetic Radiation, Department of Occupational Health, Ministry of Education, Third Military Medical University, Chongqing, China
| | - Qinlong Ma
- Key Laboratory of Medical Protection for Electromagnetic Radiation, Department of Occupational Health, Ministry of Education, Third Military Medical University, Chongqing, China
| | - Huifeng Pi
- Key Laboratory of Medical Protection for Electromagnetic Radiation, Department of Occupational Health, Ministry of Education, Third Military Medical University, Chongqing, China
| | - Mindi He
- Key Laboratory of Medical Protection for Electromagnetic Radiation, Department of Occupational Health, Ministry of Education, Third Military Medical University, Chongqing, China
| | - Yonghui Lu
- Key Laboratory of Medical Protection for Electromagnetic Radiation, Department of Occupational Health, Ministry of Education, Third Military Medical University, Chongqing, China
| | - Peng Gao
- Key Laboratory of Medical Protection for Electromagnetic Radiation, Department of Occupational Health, Ministry of Education, Third Military Medical University, Chongqing, China
| | - Chao Zhou
- Key Laboratory of Medical Protection for Electromagnetic Radiation, Department of Occupational Health, Ministry of Education, Third Military Medical University, Chongqing, China
| | - Zhixin He
- Key Laboratory of Medical Protection for Electromagnetic Radiation, Department of Occupational Health, Ministry of Education, Third Military Medical University, Chongqing, China
| | - Yanwen Zhang
- Key Laboratory of Medical Protection for Electromagnetic Radiation, Department of Occupational Health, Ministry of Education, Third Military Medical University, Chongqing, China
| | - Zhengping Yu
- Key Laboratory of Medical Protection for Electromagnetic Radiation, Department of Occupational Health, Ministry of Education, Third Military Medical University, Chongqing, China
| | - Lei Zhang
- Key Laboratory of Medical Protection for Electromagnetic Radiation, Department of Occupational Health, Ministry of Education, Third Military Medical University, Chongqing, China
| |
Collapse
|
43
|
Maneshi P, Mason J, Dongre M, Öhlund D. Targeting Tumor-Stromal Interactions in Pancreatic Cancer: Impact of Collagens and Mechanical Traits. Front Cell Dev Biol 2021; 9:787485. [PMID: 34901028 PMCID: PMC8656238 DOI: 10.3389/fcell.2021.787485] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/01/2021] [Indexed: 01/18/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has one of the worst outcomes among cancers with a 5-years survival rate of below 10%. This is a result of late diagnosis and the lack of effective treatments. The tumor is characterized by a highly fibrotic stroma containing distinct cellular components, embedded within an extracellular matrix (ECM). This ECM-abundant tumor microenvironment (TME) in PDAC plays a pivotal role in tumor progression and resistance to treatment. Cancer-associated fibroblasts (CAFs), being a dominant cell type of the stroma, are in fact functionally heterogeneous populations of cells within the TME. Certain subtypes of CAFs are the main producer of the ECM components of the stroma, with the most abundant one being the collagen family of proteins. Collagens are large macromolecules that upon deposition into the ECM form supramolecular fibrillar structures which provide a mechanical framework to the TME. They not only bring structure to the tissue by being the main structural proteins but also contain binding domains that interact with surface receptors on the cancer cells. These interactions can induce various responses in the cancer cells and activate signaling pathways leading to epithelial-to-mesenchymal transition (EMT) and ultimately metastasis. In addition, collagens are one of the main contributors to building up mechanical forces in the tumor. These forces influence the signaling pathways that are involved in cell motility and tumor progression and affect tumor microstructure and tissue stiffness by exerting solid stress and interstitial fluid pressure on the cells. Taken together, the TME is subjected to various types of mechanical forces and interactions that affect tumor progression, metastasis, and drug response. In this review article, we aim to summarize and contextualize the recent knowledge of components of the PDAC stroma, especially the role of different collagens and mechanical traits on tumor progression. We furthermore discuss different experimental models available for studying tumor-stromal interactions and finally discuss potential therapeutic targets within the stroma.
Collapse
Affiliation(s)
- Parniyan Maneshi
- Department of Radiation Sciences, Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - James Mason
- Department of Radiation Sciences, Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Mitesh Dongre
- Department of Radiation Sciences, Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Daniel Öhlund
- Department of Radiation Sciences, Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| |
Collapse
|
44
|
Sanvee GM, Hitzfeld L, Bouitbir J, Krähenbühl S. mTORC2 is an important target for simvastatin-associated toxicity in C2C12 cells and mouse skeletal muscle - Roles of Rap1 geranylgeranylation and mitochondrial dysfunction. Biochem Pharmacol 2021; 192:114750. [PMID: 34461118 DOI: 10.1016/j.bcp.2021.114750] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 12/25/2022]
Abstract
Statins decrease the serum LDL-cholesterol concentration and reduce the risk for cardiovascular diseases but can cause myopathy, which may be related to mTORC inhibition. In the current study, we investigated which mTORC is inhibited by simvastatin and by which mechanisms. In C2C12 myoblasts and myotubes and mouse gastrocnemius, simvastatin was cytotoxic and inhibited S6rp and Akt Ser473 phosphorylation, indicating inhibition of mTORC1 and mTORC2, respectively. In contrast to simvastatin, the mTORC1 inhibitor rapamycin did not inhibit mTORC2 activity and was not cytotoxic. Like simvastatin, knock-down of Rictor, an essential component of mTORC2, impaired Akt Ser473 and S6rp phosphorylation and was cytotoxic for C2C12 myoblasts, suggesting that mTORC2 inhibition is an important myotoxic mechanism. The investigation of the mechanism of mTORC2 inhibition showed that simvastatin impaired Ras farnesylation, which was prevented by farnesol but without restoring mTORC2 activity. In comparison, Rap1 knock-down reduced mTORC2 activity and was cytotoxic for C2C12 myoblasts. Simvastatin impaired Rap1 geranylgeranylation and function, which was prevented by geranylgeraniol. In addition, simvastatin and the complex III inhibitor antimycin A caused mitochondrial superoxide accumulation and impaired the activity of mTORC2, which could partially be prevented by the antioxidant MitoTEMPO. In conclusion, mTORC2 inhibition is an important mechanism of simvastatin-induced myotoxicity. Simvastatin inhibits mTORC2 by impairing geranylgeranylation of Rap1 and by inducing mitochondrial dysfunction.
Collapse
Affiliation(s)
- Gerda M Sanvee
- Division of Clinical Pharmacology & Toxicology, University Hospital, Basel, Switzerland; Department of Biomedicine, University of Basel, Switzerland
| | - Leonie Hitzfeld
- Division of Clinical Pharmacology & Toxicology, University Hospital, Basel, Switzerland
| | - Jamal Bouitbir
- Division of Clinical Pharmacology & Toxicology, University Hospital, Basel, Switzerland; Department of Biomedicine, University of Basel, Switzerland; Division of Molecular and Systemic Toxicology, Department of Pharmaceutical Sciences, University of Basel, Switzerland; Swiss Centre for Applied Human Research (SCAHT), Switzerland
| | - Stephan Krähenbühl
- Division of Clinical Pharmacology & Toxicology, University Hospital, Basel, Switzerland; Department of Biomedicine, University of Basel, Switzerland; Swiss Centre for Applied Human Research (SCAHT), Switzerland.
| |
Collapse
|
45
|
Chen YJ, Chang JT, You GR, Huang CY, Fan KH, Cheng AJ. Panel biomarkers associated with cancer invasion and prognostic prediction for head-neck cancer. Biomark Med 2021; 15:861-877. [PMID: 34032473 DOI: 10.2217/bmm-2021-0213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 04/28/2021] [Indexed: 11/21/2022] Open
Abstract
Aim: Cell invasion leading to metastasis is a major cause of treatment failure in head-neck cancers (HNCs). Identifying prognostic molecules associated with invasiveness is imperative for clinical applications. Materials & methods: A systemic approach was used to globally survey invasion-related genes, including transcriptomic profiling, pathway analysis, data mining and prognostic assessment using TCGA-HNSC dataset. Results: Six functional pathways and six hub molecules (LAMA3, LAMC2, THBS1, IGF1R, PDGFB and TGFβ1) were identified that significantly contributed to cell invasion, leading to poor survival in HNC patients. Combinations of multiple biomarkers substantially increased the probability of accurately predicting prognosis. Conclusion: Our six defined invasion-related molecules may be used as a panel signature in precision medicine for prognostic indicators or molecular therapeutic targets for HNC.
Collapse
Affiliation(s)
- Yin-Ju Chen
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei, 11031, Taiwan
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Joseph T Chang
- Department of Radiation Oncology, Chang Gung Memorial Hospital-Linkou, Taoyuan, 33333, Taiwan
- Department of Medical School, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
| | - Guo-Rung You
- Department of Medical Biotechnology & Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
| | - Chun-Yu Huang
- School of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan
| | - Kang-Hsing Fan
- Department of Radiation Oncology, Chang Gung Memorial Hospital-Linkou, Taoyuan, 33333, Taiwan
- Department of Radiation Oncology, New Taipei Municipal TuCheng Hospital, New Taipei City, 236017, Taiwan
| | - Ann-Joy Cheng
- Department of Radiation Oncology, Chang Gung Memorial Hospital-Linkou, Taoyuan, 33333, Taiwan
- Department of Medical Biotechnology & Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
| |
Collapse
|
46
|
You GR, Chang JT, Li YL, Chen YJ, Huang YC, Fan KH, Chen YC, Kang CJ, Cheng AJ. Molecular Interplays Between Cell Invasion and Radioresistance That Lead to Poor Prognosis in Head-Neck Cancer. Front Oncol 2021; 11:681717. [PMID: 34307149 PMCID: PMC8299304 DOI: 10.3389/fonc.2021.681717] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 06/28/2021] [Indexed: 12/13/2022] Open
Abstract
Background Cancer metastasis and recurrence after radiotherapy are the significant causes of poor prognosis in head-neck cancer (HNC). Clinically, it is commonly found that patients with either condition may accompany the outcome of the other. We hypothesized that HNC cells might exhibit a cross-phenotypic attribute between cell invasion and radioresistance. To discover effective biomarkers for the intervention of aggressive cancer at one time, the potential molecules that interplay between these two phenotypes were investigated. Materials and Methods Three isogenic HNC cell sublines with high invasion or radioresistance properties were established. Transcriptomic and bioinformatic methods were used to globally assess the phenotypic-specific genes, functional pathways, and co-regulatory hub molecules. The associations of gene expressions with patient survival were analyzed by Kaplan-Meier plotter, a web-based tool, using the HNSCC dataset (n=500). The molecular and cellular techniques, including RT-qPCR, flow cytometry, cell invasion assay, and clonogenic survival assay, were applied. Results The phenotypic crosstalk between cell invasion and radioresistance was validated, as shown by the existence of mutual properties in each HNC subline. A total of 695 genes was identified in associations with these two phenotypes, including 349 upregulated and 346 downregulated in HNC cells. The focal adhesion mechanism showed the most significant pathway to co-regulate these functions. In the analysis of 20 up-regulatory genes, a general portrait of correlative expression was found between these phenotypic cells (r=0.513, p=0.021), and nine molecules exhibited significant associations with poor prognosis in HNC patients (HR>1, p<0.050). Three hub genes were identified (ITGA6, TGFB1, and NDRG1) that represented a signature of interplayed molecules contributing to cell invasion, radioresistance and leading to poor prognosis. The ITGA6 was demonstrated as a prominent biomarker. The expression of ITGA6 correlated with the levels of several extracellular and apoptotic/anti-apoptotic molecules. Functionally, silencing ITGA6 suppressed cell migration, invasion, and attenuated radioresistance in HNC cells. Conclusions A panel of interplay molecules was identified that contribute to cell invasion and radioresistance, leading to poor prognosis. These panel molecules, such as ITGA6, may serve as predictive markers of radioresistance, prognostic markers of metastasis, and molecular therapeutic targets for refractory HNC.
Collapse
Affiliation(s)
- Guo-Rung You
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Joseph T Chang
- Department of Radiation Oncology, Chang Gung Memorial Hospital-Linkou, Taoyuan, Taiwan.,School of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yan-Liang Li
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yin-Ju Chen
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan.,International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Yu-Chen Huang
- Department of Oral Maxillofacial Surgery, Chang Gung Memorial Hospital-Linkou, Taoyuan, Taiwan
| | - Kang-Hsing Fan
- Department of Radiation Oncology, Chang Gung Memorial Hospital-Linkou, Taoyuan, Taiwan.,Department of Radiation Oncology, New Taipei Municipal TuCheng Hospital, New Taipei City, Taiwan.,Department of Medical Imaging and Radiological Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yen-Chao Chen
- Department of Radiation Oncology, Chang Gung Memorial Hospital-Keelung, Keelung, Taiwan
| | - Chung-Jan Kang
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Otorhinolaryngology, Chang Gung Memorial Hospital-LinKou, Taoyuan, Taiwan
| | - Ann-Joy Cheng
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Radiation Oncology, Chang Gung Memorial Hospital-Linkou, Taoyuan, Taiwan.,Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
47
|
Zhang N, Wang G, Sun G. Actin-binding protein, IQGAP1, regulates LPS-induced RPMVECs hyperpermeability and ICAM-1 upregulation via Rap1/Src signalling pathway. Cell Signal 2021; 85:110067. [PMID: 34147590 DOI: 10.1016/j.cellsig.2021.110067] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/11/2021] [Accepted: 06/15/2021] [Indexed: 11/30/2022]
Abstract
Pulmonary microvascular barrier dysfunction is a hallmark feature of acute lung injury (ALI). IQGAP1 is a ubiquitously expressed scaffolding protein known to regulate cancer metastasis, angiogenesis, and barrier stability. However, the function of IQGAP1 in lipopolysaccharide (LPS)-induced microvascular endothelial hyperpermeability remains poorly understood. In the present study, we demonstrated that IQGAP1 was markedly upregulated in LPS-induced ALI models and rat pulmonary microvascular endothelial cells (RPMVECs). Lentivirus-mediated knockdown of IQGAP1 significantly attenuated the formation of actin stress fibers, phosphorylation of myosin light chain (MLC), and disruption of VE-cadherin, thereby protecting the RPMVECs barrier failure from LPS damage. In addition, IQGAP1 depletion reduced the reactive oxygen species (ROS)-mediated increase in intracellular adhesion molecule-1 (ICAM-1) in RPMVECs stimulated with LPS. Mechanistically, we found that the upregulation of IQGAP1 affected the activity of Rap1 and the downstream phosphorylation of Src. In conclusion, these findings reveal an essential mechanism by which increased IQGAP1 in LPS-treated RPMVECs promotes barrier dysfunction and ICAM-1 upregulation, at least in part by regulating Rap1/Src signalling, indicating that IQGAP1 may be a potential therapeutic target to prevent endothelial hyperpermeability and inflammation in ALI.
Collapse
Affiliation(s)
- Na Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, People's Republic of China
| | - Gang Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, People's Republic of China
| | - Gengyun Sun
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, People's Republic of China.
| |
Collapse
|
48
|
Vermot A, Petit-Härtlein I, Smith SME, Fieschi F. NADPH Oxidases (NOX): An Overview from Discovery, Molecular Mechanisms to Physiology and Pathology. Antioxidants (Basel) 2021; 10:890. [PMID: 34205998 PMCID: PMC8228183 DOI: 10.3390/antiox10060890] [Citation(s) in RCA: 315] [Impact Index Per Article: 78.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/21/2021] [Accepted: 05/26/2021] [Indexed: 01/17/2023] Open
Abstract
The reactive oxygen species (ROS)-producing enzyme NADPH oxidase (NOX) was first identified in the membrane of phagocytic cells. For many years, its only known role was in immune defense, where its ROS production leads to the destruction of pathogens by the immune cells. NOX from phagocytes catalyzes, via one-electron trans-membrane transfer to molecular oxygen, the production of the superoxide anion. Over the years, six human homologs of the catalytic subunit of the phagocyte NADPH oxidase were found: NOX1, NOX3, NOX4, NOX5, DUOX1, and DUOX2. Together with the NOX2/gp91phox component present in the phagocyte NADPH oxidase assembly itself, the homologs are now referred to as the NOX family of NADPH oxidases. NOX are complex multidomain proteins with varying requirements for assembly with combinations of other proteins for activity. The recent structural insights acquired on both prokaryotic and eukaryotic NOX open new perspectives for the understanding of the molecular mechanisms inherent to NOX regulation and ROS production (superoxide or hydrogen peroxide). This new structural information will certainly inform new investigations of human disease. As specialized ROS producers, NOX enzymes participate in numerous crucial physiological processes, including host defense, the post-translational processing of proteins, cellular signaling, regulation of gene expression, and cell differentiation. These diversities of physiological context will be discussed in this review. We also discuss NOX misregulation, which can contribute to a wide range of severe pathologies, such as atherosclerosis, hypertension, diabetic nephropathy, lung fibrosis, cancer, or neurodegenerative diseases, giving this family of membrane proteins a strong therapeutic interest.
Collapse
Affiliation(s)
- Annelise Vermot
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 38000 Grenoble, France; (A.V.); (I.P.-H.)
| | - Isabelle Petit-Härtlein
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 38000 Grenoble, France; (A.V.); (I.P.-H.)
| | - Susan M. E. Smith
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA 30144, USA;
| | - Franck Fieschi
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 38000 Grenoble, France; (A.V.); (I.P.-H.)
| |
Collapse
|
49
|
Rap1GAP Mediates Angiotensin II-Induced Cardiomyocyte Hypertrophy by Inhibiting Autophagy and Increasing Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:7848027. [PMID: 33936386 PMCID: PMC8062190 DOI: 10.1155/2021/7848027] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 03/05/2021] [Accepted: 03/18/2021] [Indexed: 12/18/2022]
Abstract
Abnormal autophagy and oxidative stress contribute to angiotensin II- (Ang II-) induced cardiac hypertrophy and heart failure. We previously showed that Ang II increased Rap1GAP gene expression in cardiomyocytes associated with hypertrophy and autophagy disorders. Using real-time PCR and Western blot, we found that Rap1GAP expression was increased in the heart of Sprague Dawley (SD) rats infused by Ang II compared with saline infusion and in Ang II vs. vehicle-treated rat neonatal cardiomyocytes. Overexpression of Rap1GAP in cultured cardiomyocytes exacerbated Ang II-induced cardiomyocyte hypertrophy, reactive oxygen species (ROS) generation, and cell apoptosis and inhibited autophagy. The increased oxidative stress caused by Rap1GAP overexpression was inhibited by the treatment of autophagy agonists. Knockdown of Rap1GAP by siRNA markedly attenuated Ang II-induced cardiomyocyte hypertrophy and oxidative stress and enhanced autophagy. The AMPK/AKT/mTOR signaling pathway was inhibited by overexpression of Rap1GAP and activated by the knockdown of Rap1GAP. These results show that Rap1GAP-mediated pathway might be a new mechanism of Ang II-induced cardiomyocyte hypertrophy, which could be a potential target for the future treatment of cardiac hypertrophy and heart failure.
Collapse
|
50
|
Kútna V, O'Leary VB, Newman E, Hoschl C, Ovsepian SV. Revisiting Brain Tuberous Sclerosis Complex in Rat and Human: Shared Molecular and Cellular Pathology Leads to Distinct Neurophysiological and Behavioral Phenotypes. Neurotherapeutics 2021; 18:845-858. [PMID: 33398801 PMCID: PMC8423952 DOI: 10.1007/s13311-020-01000-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2020] [Indexed: 12/27/2022] Open
Abstract
Tuberous sclerosis complex (TSC) is a dominant autosomal genetic disorder caused by loss-of-function mutations in TSC1 and TSC2, which lead to constitutive activation of the mammalian target of rapamycin C1 (mTORC1) with its decoupling from regulatory inputs. Because mTORC1 integrates an array of molecular signals controlling protein synthesis and energy metabolism, its unrestrained activation inflates cell growth and division, resulting in the development of benign tumors in the brain and other organs. In humans, brain malformations typically manifest through a range of neuropsychiatric symptoms, among which mental retardation, intellectual disabilities with signs of autism, and refractory seizures, which are the most prominent. TSC in the rat brain presents the first-rate approximation of cellular and molecular pathology of the human brain, showing many instructive characteristics. Nevertheless, the developmental profile and distribution of lesions in the rat brain, with neurophysiological and behavioral manifestation, deviate considerably from humans, raising numerous research and translational questions. In this study, we revisit brain TSC in human and Eker rats to relate their histopathological, electrophysiological, and neurobehavioral characteristics. We discuss shared and distinct aspects of the pathology and consider factors contributing to phenotypic discrepancies. Given the shared genetic cause and molecular pathology, phenotypic deviations suggest an incomplete understanding of the disease. Narrowing the knowledge gap in the future should not only improve the characterization of the TSC rat model but also explain considerable variability in the clinical manifestation of the disease in humans.
Collapse
Affiliation(s)
- Viera Kútna
- Department of Experimental Neurobiology, National Institute of Mental Health, Topolová 748, 250 67, Klecany, Czech Republic.
| | - Valerie B O'Leary
- Department of Medical Genetics, Third Faculty of Medicine of Charles University, Ruská 87, 100 00, Prague, Czech Republic
| | - Ehren Newman
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Cyril Hoschl
- Department of Experimental Neurobiology, National Institute of Mental Health, Topolová 748, 250 67, Klecany, Czech Republic
- Department of Psychiatry and Medical Psychology, Third Faculty of Medicine of Charles University, Ruská 87, 100 00, Prague, Czech Republic
| | - Saak V Ovsepian
- Department of Experimental Neurobiology, National Institute of Mental Health, Topolová 748, 250 67, Klecany, Czech Republic.
- Department of Psychiatry and Medical Psychology, Third Faculty of Medicine of Charles University, Ruská 87, 100 00, Prague, Czech Republic.
| |
Collapse
|