1
|
Thabet EH, Khalil NA, Essawy MM, Harby SA, Solaiman AA, El Gazaerly HM, Khalifa YH. MiRNA-21-loaded chitosan nanoparticles ameliorate pancreatic apoptosis and oxidative stress in diabetic rats. Life Sci 2025; 366-367:123471. [PMID: 39956187 DOI: 10.1016/j.lfs.2025.123471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 02/09/2025] [Accepted: 02/13/2025] [Indexed: 02/18/2025]
Abstract
BACKGROUND Accelerated Pancreatic β-cell apoptosis and oxidative stress are the mainstays of type-1 diabetes. MicroRNA-21's (miRNA-21) role in regulating pancreatic β-cell function remains indefinable. MATERIAL AND METHODS Five groups of rats were used in this study (healthy controls (Ia), controls that received only chitosan (CS) nanoparticles (NPs)(Ib), streptozotocin (STZ)-induced diabetics rats (II),STZ-induced diabetic rats that received only CS-NPs(III), and STZ-induced diabetic rats treated with mi-RNA-21-CS-NPs(IV). Sera were collected for measurement of fasting blood glucose levels (FBG), insulin, oxidative stress, and intraperitoneal glucose intolerance tests. Pancreatic tissue was collected after sacrifice partly for histological examination and for oxidative stress assessment and evaluation of PTEN/ AKT using qRT-PCR. KEY FINDINGS We showed over-expression of cleaved-caspase-3 indicating accelerated apoptosis in the β-cell of STZ-induced diabetic rats. Apoptosis was significantly ameliorated by miRNA-21-CS. MiRNA-21-CS-NPs faithfully restored serum fasting insulin, and FBG, and reduced serum and pancreatic oxidative stress markers while enhancing the total antioxidant capacity. Histological examination revealed that miRNA-21 restored healthy β-cell architecture, decreased cleaved-caspase-3, and increased insulin secretion. Transmission electron microscopy revealed increased mitochondrial circularity that significantly correlated with an exaggerated oxidative stress profile as shown by high serum and pancreatic malondialdehyde (MDA), low glutathione peroxidase, and total antioxidant capacity in STZ-induced diabetes. This oxidative profile was reversed using miRNA-21-CS-NPs. Mi-RNA-21 therapy downregulated PTEN but increased AKT and pAKT expression. Altogether, we show that miRNA-21 restored normal islet β-cell structure and insulin secretion through PTEN inhibition. SIGNIFICANCE miRNA-21- CS-NPs are promising targeted therapeutics that may effectively decrease the global burden of diabetes.
Collapse
Affiliation(s)
- Eman H Thabet
- Department of Medical Physiology, Faculty of Medicine, University of Alexandria, 21500 Alexandria, Egypt; Center of Excellence for Research in Regenerative Medicine and Applications (CERRMA), Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt.
| | - Nehal A Khalil
- Department of Medical Biochemistry, Faculty of Medicine, University of Alexandria, 21500 Alexandria, Egypt; Center of Excellence for Research in Regenerative Medicine and Applications (CERRMA), Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Marwa M Essawy
- Oral Pathology Department, Faculty of Dentistry, Alexandria University, 21500 Alexandria, Egypt; Center of Excellence for Research in Regenerative Medicine and Applications (CERRMA), Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Sahar A Harby
- Department of Clinical Pharmacology, Faculty of Medicine, University of Alexandria, 21500 Alexandria, Egypt
| | - Amany A Solaiman
- Department of Histology and Cell Biology, Faculty of Medicine, University of Alexandria, 21500 Alexandria, Egypt
| | - Hanaa M El Gazaerly
- Department of Oral Pathology, Faculty of Dentistry, Tanta University, Tanta 31527, Egypt
| | - Yassmin H Khalifa
- Department of Histology and Cell Biology, Faculty of Medicine, University of Alexandria, 21500 Alexandria, Egypt
| |
Collapse
|
2
|
Premeti K, Tsipa D, Nadalis AE, Papanikolaou MG, Syropoulou V, Karagkiozeli KD, Aggelis G, Iordanidou E, Labrakakis C, Pappas P, Keramidas AD, Antoniou K, Doulias PT, Kabanos TA, Leondaritis G. First generation vanadium-based PTEN inhibitors: Comparative study in vitro and in vivo and identification of a novel mechanism of action. Biochem Pharmacol 2025; 233:116756. [PMID: 39824468 DOI: 10.1016/j.bcp.2025.116756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 11/29/2024] [Accepted: 01/13/2025] [Indexed: 01/20/2025]
Abstract
PTEN, a tumor suppressor phosphatase, regulates cellular functions by antagonizing the growth promoting PI3K/Akt/mTOR pathway through the dephosphorylation of the second messenger PIP3. Many preclinical cellular and animal studies have used PTEN inhibitors to highlight specific disease contexts where acute activation of PI3K/Akt/mTOR pathway might offer therapeutic advantages. In the present study we have re-evaluated first-generation PTEN inhibitors, including established bisperoxo-vanadium(V) complexes (bpVs). In vitro, all compounds tested inhibited PTEN with IC50 values between 0.2-0.8 μM, although their activity diminished under reducing conditions. bpV(phen) and bpV(HΟpic) significantly increased pSer473Akt levels in PTEN wild-type cells while bpV(phen) induced phosphorylation in PTEN null cells upon re-expression of functional PTEN. bpV(ΗΟpic) was less specific since it also triggered PTEN-independent Erk1/2 phosphorylation. In vivo, bpV(phen) administration in Wistar rats enhanced pS6 levels in kidney and liver tissues, but not in several CNS tissues, and led to reduced locomotion and exploratory behaviour in the open field test. The consensus mechanism of action of first generation PTEN inhibitors appears to be oxidative inhibition, however bpV(phen) does not induce oxidation of cellular endogenous PTEN. Instead, our findings suggest that the inhibition of PTEN by bpV(phen) in cells and in vivo may proceed through a mechanism involving non-specific S-nitrosylation of PTEN. Our study highlights the complexity of PTEN inhibition by first-generation compounds and their limitations, such as low specificity, adverse effects and non-specific mechanisms of action, and emphasizes the need for developing more selective and potent PTEN inhibitors with improved efficacy and well-defined mechanisms of actions.
Collapse
Affiliation(s)
- Kyriaki Premeti
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Dimitra Tsipa
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Antonios E Nadalis
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Michael G Papanikolaou
- Department of Chemistry, University of Ioannina, Ioannina 45110, Greece; Department of Chemistry, University of Cyprus, Nicosia 2109, Cyprus
| | - Vasiliki Syropoulou
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Konstantina-Danai Karagkiozeli
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - George Aggelis
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Eleni Iordanidou
- Department of Chemistry, University of Ioannina, Ioannina 45110, Greece
| | - Charalampos Labrakakis
- Department of Biological Applications and Technology, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; Institute of Biosciences, University Research Center Ioannina, University of Ioannina, Ioannina, Greece
| | - Periklis Pappas
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | | | - Katerina Antoniou
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; Institute of Biosciences, University Research Center Ioannina, University of Ioannina, Ioannina, Greece
| | - Paschalis-Thomas Doulias
- Department of Chemistry, University of Ioannina, Ioannina 45110, Greece; Institute of Biosciences, University Research Center Ioannina, University of Ioannina, Ioannina, Greece
| | | | - George Leondaritis
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; Institute of Biosciences, University Research Center Ioannina, University of Ioannina, Ioannina, Greece.
| |
Collapse
|
3
|
Jalise SZ, Habibi S, Fath-Bayati L, Habibi MA, Ababzadeh S, Hosseinzadeh F. Role and Interplay of Different Signaling Pathways Involved in Sciatic Nerve Regeneration. J Mol Neurosci 2024; 74:108. [PMID: 39531101 DOI: 10.1007/s12031-024-02286-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024]
Abstract
Regeneration of the sciatic nerve is a sophisticated process that involves the interplay of several signaling pathways that orchestrate the cellular responses critical to regeneration. Among the key pathways are the mitogen-activated protein kinase (MAPK), phosphatidylinositol 3-kinase (PI3K)/AKT, cyclic adenosine monophosphate (cAMP), and Janus kinase/signal transducers and transcription activators (JAK/STAT) pathways. In particular, the cAMP pathway modulates neuronal survival and axonal regrowth. It influences various cellular behaviors and gene expression that are essential for nerve regeneration. MAPK is indispensable for Schwann cell differentiation and myelination, whereas PI3K/AKT is integral to the transcription, translation, and cell survival processes that are vital for nerve regeneration. Furthermore, GTP-binding proteins, including those of the Ras homolog gene family (Rho), regulate neural cell adhesion, migration, and survival. Notch signaling also appears to be effective in the early stages of nerve regeneration and in preventing skeletal muscle fibrosis after injury. Understanding the intricate mechanisms and interactions of these pathways is vital for the development of effective therapeutic strategies for sciatic nerve injuries. This review underscores the need for further research to fill existing knowledge gaps and improve therapeutic outcomes.
Collapse
Affiliation(s)
- Saeedeh Zare Jalise
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran
- Cellular and Molecular Research Centre, Qom University of Medical Sciences, Qom, Iran
| | - Sina Habibi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Leyla Fath-Bayati
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran
- Cellular and Molecular Research Centre, Qom University of Medical Sciences, Qom, Iran
| | - Mohammad Amin Habibi
- Clinical Research Development Center, Shahid Beheshti Hospital, Qom University of Medical Sciences, Qom, Iran
| | - Shima Ababzadeh
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran.
- Cellular and Molecular Research Centre, Qom University of Medical Sciences, Qom, Iran.
| | - Faezeh Hosseinzadeh
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran.
- Cellular and Molecular Research Centre, Qom University of Medical Sciences, Qom, Iran.
- Clinical Trial Center, Qom University of Medical Sciences, Qom, Iran.
| |
Collapse
|
4
|
Zhu AZ, Ma Z, Wolff EV, Lin Z, Gao ZJ, Li X, Du W. HES1 is required for mouse fetal hematopoiesis. Stem Cell Res Ther 2024; 15:235. [PMID: 39075526 PMCID: PMC11287931 DOI: 10.1186/s13287-024-03836-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 07/06/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND Hematopoiesis in mammal is a complex and highly regulated process in which hematopoietic stem cells (HSCs) give rise to all types of differentiated blood cells. Previous studies have shown that hairy and enhancer of split (HES) repressors are essential regulators of adult HSC development downstream of Notch signaling. METHODS In this study, we investigated the role of HES1, a member of HES family, in fetal hematopoiesis using an embryonic hematopoietic specific Hes1 conditional knockout mouse model by using phenotypic flow cytometry, histopathology analysis, and functional in vitro colony forming unit (CFU) assay and in vivo bone marrow transplant (BMT) assay. RESULTS We found that loss of Hes1 in early embryonic stage leads to smaller embryos and fetal livers, decreases hematopoietic stem progenitor cell (HSPC) pool, results in defective multi-lineage differentiation. Functionally, fetal hematopoietic cells deficient for Hes1 exhibit reduced in vitro progenitor activity and compromised in vivo repopulation capacity in the transplanted recipients. Further analysis shows that fetal hematopoiesis defects in Hes1fl/flFlt3Cre embryos are resulted from decreased proliferation and elevated apoptosis, associated with de-repressed HES1 targets, p27 and PTEN in Hes1-KO fetal HSPCs. Finally, pharmacological inhibition of p27 or PTEN improves fetal HSPCs function both in vitro and in vivo. CONCLUSION Together, our findings reveal a previously unappreciated role for HES1 in regulating fetal hematopoiesis, and provide new insight into the differences between fetal and adult HSC maintenance.
Collapse
Affiliation(s)
- Anthony Z Zhu
- Division of Hematology and Oncology, University of Pittsburgh School of Medicine, 5117 Center Ave, Pittsburgh, PA, 15213, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
| | - Zhilin Ma
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | - Emily V Wolff
- Division of Hematology and Oncology, University of Pittsburgh School of Medicine, 5117 Center Ave, Pittsburgh, PA, 15213, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
| | - Zichen Lin
- Master of Science in Medical Science, Boston University School of Medicine Graduate Master Program, Boston, MA, USA
| | - Zhenxia J Gao
- Division of Hematology and Oncology, University of Pittsburgh School of Medicine, 5117 Center Ave, Pittsburgh, PA, 15213, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
| | - Xue Li
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China
| | - Wei Du
- Division of Hematology and Oncology, University of Pittsburgh School of Medicine, 5117 Center Ave, Pittsburgh, PA, 15213, USA.
- UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
5
|
Trinh VH, Choi JM, Nguyen Huu T, Sah DK, Yoon HJ, Park SC, Jung YS, Ahn YK, Lee KH, Lee SR. Redox Regulation of Phosphatase and Tensin Homolog by Bicarbonate and Hydrogen Peroxide: Implication of Peroxymonocarbonate in Cell Signaling. Antioxidants (Basel) 2024; 13:473. [PMID: 38671920 PMCID: PMC11047460 DOI: 10.3390/antiox13040473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 04/05/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
Phosphatase and tensin homolog (PTEN) is a negative regulator of the phosphoinositide 3-kinases/protein kinase B (PI3K/AKT) signaling pathway. Notably, its active site contains a cysteine residue that is susceptible to oxidation by hydrogen peroxide (H2O2). This oxidation inhibits the phosphatase function of PTEN, critically contributing to the activation of the PI3K/AKT pathway. Upon the stimulation of cell surface receptors, the activity of NADPH oxidase (NOX) generates a transient amount of H2O2, serving as a mediator in this pathway by oxidizing PTEN. The mechanism underlying this oxidation, occurring despite the presence of highly efficient and abundant cellular oxidant-protecting and reducing systems, continues to pose a perplexing conundrum. Here, we demonstrate that the presence of bicarbonate (HCO3-) promoted the rate of H2O2-mediated PTEN oxidation, probably through the formation of peroxymonocarbonate (HCO4-), and consequently potentiated the phosphorylation of AKT. Acetazolamide (ATZ), a carbonic anhydrase (CA) inhibitor, was shown to diminish the oxidation of PTEN. Thus, CA can also be considered as a modulator in this context. In essence, our findings consolidate the crucial role of HCO3- in the redox regulation of PTEN by H2O2, leading to the presumption that HCO4- is a signaling molecule during cellular physiological processes.
Collapse
Affiliation(s)
- Vu Hoang Trinh
- Department of Biochemistry, Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 501190, Republic of Korea; (V.H.T.); (T.N.H.); (D.K.S.); (H.-J.Y.)
- Department of Oncology, Department of Medical Sciences, Pham Ngoc Thach University of Medicine, Ho Chi Minh City 700000, Vietnam
| | - Jin-Myung Choi
- Luxanima Inc., Room 102, 12-55, Sandan-gil, Hwasun-eup, Hwasun-gun 58128, Republic of Korea;
| | - Thang Nguyen Huu
- Department of Biochemistry, Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 501190, Republic of Korea; (V.H.T.); (T.N.H.); (D.K.S.); (H.-J.Y.)
| | - Dhiraj Kumar Sah
- Department of Biochemistry, Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 501190, Republic of Korea; (V.H.T.); (T.N.H.); (D.K.S.); (H.-J.Y.)
| | - Hyun-Joong Yoon
- Department of Biochemistry, Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 501190, Republic of Korea; (V.H.T.); (T.N.H.); (D.K.S.); (H.-J.Y.)
| | - Sang-Chul Park
- The Future Life & Society Research Center, Advanced Institute of Aging Science, Chonnam National University, Gwangju 61469, Republic of Korea;
| | - Yu-Seok Jung
- Chonnam National University Medical School, Gwangju 501190, Republic of Korea;
| | - Young-Keun Ahn
- Department of Cardiology, Chonnam National University Hospital, Gwangju 61469, Republic of Korea;
| | - Kun-Ho Lee
- Department of Biomedical Science, Chosun University, Gwangju 61452, Republic of Korea;
- Department of Neural Development and Disease, Korea Brain Research Institute, Daegu 41062, Republic of Korea
| | - Seung-Rock Lee
- Department of Biochemistry, Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 501190, Republic of Korea; (V.H.T.); (T.N.H.); (D.K.S.); (H.-J.Y.)
| |
Collapse
|
6
|
Monteiro MM, Amorim Dos Santos J, Paiva Barbosa V, Rezende TMB, Guerra ENS. Photobiomodulation effects on fibroblasts and keratinocytes after ionizing radiation and bacterial stimulus. Arch Oral Biol 2024; 159:105874. [PMID: 38147800 DOI: 10.1016/j.archoralbio.2023.105874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/02/2023] [Accepted: 12/13/2023] [Indexed: 12/28/2023]
Abstract
OBJECTIVE Photobiomodulation therapy (PBMT) has proven to reduce inflammation and pain and increase wound healing. Thus, the aim of this study was to analyze the effects of PBMT parameters on migration, proliferation, and gene expression after ionizing radiation and bacterial-induced stress in an in vitro study. DESIGN Keratinocytes (HaCaT) and Fibroblasts (HGFs) were grown in DMEM with 10 % fetal bovine serum until stressful condition induction with lipopolysaccharide (LPS) of Escherichia coli (1 µg/mL), Porphyromonas gingivalis protein extract (5 µg/mL) and ionizing radiation (8 Gy). Low-laser irradiation (660 nm, 30 mW) was carried out in four sessions, with 6 h intervals, and energy density of 2, 3, 4, and 5 J/cm². Scratch assays, immunofluorescence, and RT-qPCR were performed. RESULTS Treated fibroblasts and keratinocytes showed significant response in proliferation and migration after scratch assays (p < 0.05). Higher expressions of α-SMA in fibroblasts and F-actin in keratinocytes were observed in cells subjected to 3 J/cm². PI3K-pathway genes expression tended to enhance in fibroblasts, presenting a higher relative expression when compared to keratinocytes. In keratinocytes, PBMT groups demonstrated deregulated expression for all inflammatory cytokines' genes tested while fibroblasts presented a tendency to enhance those genes expression in a dose dependent way. CONCLUSIONS The present study showed that delivering 660 nm, 30 mW was effective to stimulate cell migration, proliferation and to accelerate wound healing. PBMT can modulate cytokines and pathways involved in wound repair. The different energy densities delivering distinct responses in vitro highlights that understanding laser parameters is fundamental to improve treatment strategies.
Collapse
Affiliation(s)
- Mylene Martins Monteiro
- University of Brasília, Brasília, Laboratory of Oral Histopathology, Health Sciences Faculty, Brazil
| | - Juliana Amorim Dos Santos
- University of Brasília, Brasília, Laboratory of Oral Histopathology, Health Sciences Faculty, Brazil
| | - Victor Paiva Barbosa
- University of Brasília, Brasília, Laboratory of Oral Histopathology, Health Sciences Faculty, Brazil
| | - Taia Maria Berto Rezende
- University of Brasília, Brasília, Laboratory of Oral Histopathology, Health Sciences Faculty, Brazil
| | - Eliete Neves Silva Guerra
- University of Brasília, Brasília, Laboratory of Oral Histopathology, Health Sciences Faculty, Brazil.
| |
Collapse
|
7
|
Gong L, Jiang S, Tian J, Li Y, Yu W, Zhang L, Xiao D. STZ-induced gestational diabetes exposure alters PTEN/AKT/mTOR-mediated autophagy signaling pathway leading to increase the risk of neonatal hypoxic-ischemic encephalopathy. Reprod Toxicol 2024; 123:S0890-6238(23)00168-5. [PMID: 38706688 PMCID: PMC11068333 DOI: 10.1016/j.reprotox.2023.108494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/23/2023] [Accepted: 10/26/2023] [Indexed: 05/07/2024]
Abstract
Exposure to gestational diabetes mellitus (GDM) during pregnancy has significant consequences for the unborn baby and newborn infant. However, whether and how GDM exposure induces the development of neonatal brain hypoxia/ischemia-sensitive phenotype and the underlying molecular mechanisms remain unclear. In this study, we used a late GDM rat model induced by administration of streptozotocin (STZ) on gestational day 12 and investigated its effects of GDM on neonatal brain development. The pregnant rats exhibited increased blood glucose levels in a dose-dependent manner after STZ administration. STZ-induced maternal hyperglycemia led to reduced blood glucose levels in neonatal offspring, resulting in growth restriction and an increased brain to body weight ratio. Importantly, GDM exposure increased susceptibility to hypoxia/ischemia (HI)-induced brain infarct sizes compared to the controls in both male and female neonatal offspring. Further molecular analysis revealed alterations in the PTEN/AKT/mTOR/autophagy signaling pathway in neonatal male offspring brains, along with increased ROS production and autophagy-related proteins (Atg5 and LC3-II). Treatment with the PTEN inhibitor bisperoxovanadate (BPV) eliminated the differences in HI-induced brain infarct sizes between the GDM-exposed and the control groups. These findings provide novel evidence of the development of a brain hypoxia/ischemia-sensitive phenotype in response to GDM exposure and highlight the role of the PTEN/AKT/mTOR/autophagy signaling pathway in this process.
Collapse
Affiliation(s)
- Lei Gong
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
- Institute of Medical Biotechnology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Siyi Jiang
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
- Department of Hematology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jia Tian
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Yong Li
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Wansu Yu
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Lubo Zhang
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Daliao Xiao
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| |
Collapse
|
8
|
Levina A, Uslan C, Murakami H, Crans DC, Lay PA. Substitution Kinetics, Albumin and Transferrin Affinities, and Hypoxia All Affect the Biological Activities of Anticancer Vanadium(V) Complexes. Inorg Chem 2023; 62:17804-17817. [PMID: 37858311 DOI: 10.1021/acs.inorgchem.3c02561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023]
Abstract
Limited stability of most transition-metal complexes in biological media has hampered their medicinal applications but also created a potential for novel cancer treatments, such as intratumoral injections of cytotoxic but short-lived anticancer drugs. Two related V(V) complexes, [VO(Hshed)(dtb)] (1) and [VO(Hshed)(cat)] (2), where H2shed = N-(salicylideneaminato)-N'-(2-hydroxyethyl)-1,2-ethanediamine, H2dtb = 3,5-di-tert-butylcatechol, and H2cat = 1,2-catechol, decomposed within minutes in cell culture medium at 310 K (t1/2 = 43 and 9 s for 1 and 2, respectively). Despite this, both complexes showed high antiproliferative activities in triple-negative human breast cancer (MDA-MB-231) cells, but the mechanisms of their activities were radically different. Complex 1 formed noncovalent adducts with human serum albumin, rapidly entered cells via passive diffusion, and was nearly as active in a short-term treatment (IC50 = 1.9 ± 0.2 μM at 30 min) compared with a long-term treatment (IC50 = 1.3 ± 0.2 μM at 72 h). The activity of 1 decreased about 20-fold after its decomposition in cell culture medium for 30 min at 310 K. Complex 2 showed similar activities (IC50 ≈ 12 μM at 72 h) in both fresh and decomposed solutions and was inactive in a short-term treatment. The activity of 2 was mainly due to the reactions among V(V) decomposition products, free catechol, and O2 in cell culture medium. As a result, the activity of 1 was less sensitive than that of 2 to the effects of hypoxic conditions that are characteristic of solid tumors and to the presence of apo-transferrin that acts as a scavenger of V(V/IV) decomposition products in blood serum. In summary, complex 1, but not 2, is a suitable candidate for further development as an anticancer drug delivered via intratumoral injections. These results demonstrate the importance of fine-tuning the ligand properties for the optimization of biological activities of metal complexes.
Collapse
Affiliation(s)
- Aviva Levina
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| | - Canan Uslan
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| | - Heide Murakami
- Department of Chemistry and the Cell and Molecular Biology Program, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Debbie C Crans
- Department of Chemistry and the Cell and Molecular Biology Program, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Peter A Lay
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
9
|
Gong GQ, Bilanges B, Allsop B, Masson GR, Roberton V, Askwith T, Oxenford S, Madsen RR, Conduit SE, Bellini D, Fitzek M, Collier M, Najam O, He Z, Wahab B, McLaughlin SH, Chan AWE, Feierberg I, Madin A, Morelli D, Bhamra A, Vinciauskaite V, Anderson KE, Surinova S, Pinotsis N, Lopez-Guadamillas E, Wilcox M, Hooper A, Patel C, Whitehead MA, Bunney TD, Stephens LR, Hawkins PT, Katan M, Yellon DM, Davidson SM, Smith DM, Phillips JB, Angell R, Williams RL, Vanhaesebroeck B. A small-molecule PI3Kα activator for cardioprotection and neuroregeneration. Nature 2023; 618:159-168. [PMID: 37225977 PMCID: PMC7614683 DOI: 10.1038/s41586-023-05972-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 03/17/2023] [Indexed: 05/26/2023]
Abstract
Harnessing the potential beneficial effects of kinase signalling through the generation of direct kinase activators remains an underexplored area of drug development1-5. This also applies to the PI3K signalling pathway, which has been extensively targeted by inhibitors for conditions with PI3K overactivation, such as cancer and immune dysregulation. Here we report the discovery of UCL-TRO-1938 (referred to as 1938 hereon), a small-molecule activator of the PI3Kα isoform, a crucial effector of growth factor signalling. 1938 allosterically activates PI3Kα through a distinct mechanism by enhancing multiple steps of the PI3Kα catalytic cycle and causes both local and global conformational changes in the PI3Kα structure. This compound is selective for PI3Kα over other PI3K isoforms and multiple protein and lipid kinases. It transiently activates PI3K signalling in all rodent and human cells tested, resulting in cellular responses such as proliferation and neurite outgrowth. In rodent models, acute treatment with 1938 provides cardioprotection from ischaemia-reperfusion injury and, after local administration, enhances nerve regeneration following nerve crush. This study identifies a chemical tool to directly probe the PI3Kα signalling pathway and a new approach to modulate PI3K activity, widening the therapeutic potential of targeting these enzymes through short-term activation for tissue protection and regeneration. Our findings illustrate the potential of activating kinases for therapeutic benefit, a currently largely untapped area of drug development.
Collapse
Affiliation(s)
- Grace Q Gong
- Cell Signalling, Cancer Institute, University College London, London, UK
| | - Benoit Bilanges
- Cell Signalling, Cancer Institute, University College London, London, UK
| | - Ben Allsop
- Drug Discovery Group, Translational Research Office, University College London, London, UK
| | - Glenn R Masson
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
- Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - Victoria Roberton
- UCL Centre for Nerve Engineering, UCL School of Pharmacy, University College London, London, UK
| | - Trevor Askwith
- Drug Discovery Group, Translational Research Office, University College London, London, UK
| | - Sally Oxenford
- Drug Discovery Group, Translational Research Office, University College London, London, UK
| | - Ralitsa R Madsen
- Cell Signalling, Cancer Institute, University College London, London, UK
| | - Sarah E Conduit
- Cell Signalling, Cancer Institute, University College London, London, UK
| | - Dom Bellini
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Martina Fitzek
- Hit Discovery, Discovery Sciences, R&D, AstraZeneca, Alderley Park, Macclesfield, UK
| | - Matt Collier
- Hit Discovery, Discovery Sciences, R&D, AstraZeneca, Alderley Park, Macclesfield, UK
| | - Osman Najam
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - Zhenhe He
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - Ben Wahab
- Medicines Discovery Institute, School of Biosciences, Cardiff University, Cardiff, UK
| | | | - A W Edith Chan
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | | | - Andrew Madin
- Hit Discovery, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Daniele Morelli
- Cell Signalling, Cancer Institute, University College London, London, UK
| | - Amandeep Bhamra
- Proteomics Research Translational Technology Platform, Cancer Institute, University College London, London, UK
| | - Vanesa Vinciauskaite
- Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee, UK
| | | | - Silvia Surinova
- Proteomics Research Translational Technology Platform, Cancer Institute, University College London, London, UK
| | - Nikos Pinotsis
- Institute of Structural and Molecular Biology, Birkbeck College, London, UK
| | | | - Matthew Wilcox
- UCL Centre for Nerve Engineering, UCL School of Pharmacy, University College London, London, UK
| | - Alice Hooper
- Drug Discovery Group, Translational Research Office, University College London, London, UK
| | - Chandni Patel
- Drug Discovery Group, Translational Research Office, University College London, London, UK
| | - Maria A Whitehead
- Cell Signalling, Cancer Institute, University College London, London, UK
| | - Tom D Bunney
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London, UK
| | | | | | - Matilda Katan
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London, UK
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - David M Smith
- Emerging Innovations, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - James B Phillips
- UCL Centre for Nerve Engineering, UCL School of Pharmacy, University College London, London, UK
| | - Richard Angell
- Drug Discovery Group, Translational Research Office, University College London, London, UK
- Medicines Discovery Institute, School of Biosciences, Cardiff University, Cardiff, UK
| | - Roger L Williams
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | | |
Collapse
|
10
|
Gupta S, Sharma P, Chaudhary M, Premraj S, Kaur S, Vijayan V, Arun MG, Prasad NG, Ramachandran R. Pten associates with important gene regulatory network to fine-tune Müller glia-mediated zebrafish retina regeneration. Glia 2023; 71:259-283. [PMID: 36128720 DOI: 10.1002/glia.24270] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 11/11/2022]
Abstract
Unlike mammals, zebrafish possess a remarkable ability to regenerate damaged retina after an acute injury. Retina regeneration in zebrafish involves the induction of Müller glia-derived progenitor cells (MGPCs) exhibiting stem cell-like characteristics, which are capable of restoring all retinal cell-types. The induction of MGPC through Müller glia-reprograming involves several cellular, genetic and biochemical events soon after a retinal injury. Despite the knowledge on the importance of Phosphatase and tensin homolog (Pten), which is a dual-specificity phosphatase and tumor suppressor in the maintaining of cellular homeostasis, its importance during retina regeneration remains unknown. Here, we explored the importance of Pten during zebrafish retina regeneration. The Pten gets downregulated upon retinal injury and is absent from the MGPCs, which is essential to trigger Akt-mediated cellular proliferation essential for retina regeneration. We found that the downregulation of Pten in the post-injury retina accelerates MGPCs formation, while its overexpression restricts the regenerative response. We observed that Pten regulates the proliferation of MGPCs not only through Akt pathway but also by Mmp9/Notch signaling. Mmp9-activity is essential to induce the proliferation of MGPCs in the absence of Pten. Lastly, we show that expression of Pten is fine-tuned through Mycb/histone deacetylase1 and Tgf-β signaling. The present study emphasizes on the stringent regulation of Pten and its crucial involvement during the zebrafish retina regeneration.
Collapse
Affiliation(s)
- Shivangi Gupta
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Punjab, India
| | - Poonam Sharma
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Punjab, India
| | - Mansi Chaudhary
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Punjab, India
| | - Sharanya Premraj
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Punjab, India
| | - Simran Kaur
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Punjab, India
| | - Vijithkumar Vijayan
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Punjab, India
| | - Manas Geeta Arun
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Punjab, India
| | - Nagaraj Guru Prasad
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Punjab, India
| | - Rajesh Ramachandran
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Punjab, India
| |
Collapse
|
11
|
Wu X, Wang Y, Chen H, Wang Y, Gu Y. Phosphatase and tensin homologue determine inflammatory status by differentially regulating the expression of Akt1 and Akt2 in macrophage alternative polarization of periodontitis. J Clin Periodontol 2023; 50:220-231. [PMID: 36217693 DOI: 10.1111/jcpe.13730] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/21/2022] [Accepted: 10/02/2022] [Indexed: 01/18/2023]
Abstract
AIM Macrophages are closely involved in periodontitis. However, the molecular mechanism by which macrophages influence periodontitis is not well understood. We investigated the effects of phosphatase and tensin homologue (PTEN) on macrophage polarization, the underlying mechanism and the regulatory roles in periodontium regeneration. MATERIALS AND METHODS PTEN expression in periodontitis macrophages was detected ex vivo. The effects of PTEN on macrophage polarization and the underlying mechanisms were investigated in vitro. We also analysed the ability of PTEN inhibitors to repair periodontitis in vivo in a ligature-induced mouse model of periodontitis. RESULTS Macrophage PTEN expression in periodontitis patients was significantly higher than that of controls. PTEN inhibition in macrophages induced alternative macrophage polarization, whereas PTEN overexpression facilitated classical polarization. PTEN inhibition facilitated activation of Akt1 while inhibiting expression of Akt2. Furthermore, Akt2 overexpression could rescue the effects of PTEN inhibition on NF-κB. Treatment with a PTEN inhibitor significantly attenuated the local inflammatory status and prevented alveolar bone resorption in the mouse model. CONCLUSIONS Our findings suggest that PTEN inhibition could induce alternative macrophage polarization by differentially regulating Akt1 and Akt2. This also changed a pro-inflammatory microenvironment to an anti-inflammatory environment by subsequently regulating the expression of NF-κB, thereby attenuating inflammatory alveolar bone resorption induced by ligature.
Collapse
Affiliation(s)
- Xiaowei Wu
- Department of Orthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, People's Republic of China.,Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, People's Republic of China
| | - Yidi Wang
- Department of Orthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, People's Republic of China
| | - Haotian Chen
- Department of Orthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, People's Republic of China
| | - Yixiang Wang
- Central Laboratory, Peking University School and Hospital of Stomatology, No. 22 Zhongguancun South Avenue, Haidian District, Beijing, People's Republic of China
| | - Yan Gu
- Department of Orthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, People's Republic of China
| |
Collapse
|
12
|
Kotzampasi DM, Premeti K, Papafotika A, Syropoulou V, Christoforidis S, Cournia Z, Leondaritis G. The orchestrated signaling by PI3Kα and PTEN at the membrane interface. Comput Struct Biotechnol J 2022; 20:5607-5621. [PMID: 36284707 PMCID: PMC9578963 DOI: 10.1016/j.csbj.2022.10.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 10/03/2022] [Accepted: 10/03/2022] [Indexed: 11/16/2022] Open
Abstract
The oncogene PI3Kα and the tumor suppressor PTEN represent two antagonistic enzymatic activities that regulate the interconversion of the phosphoinositide lipids PI(4,5)P2 and PI(3,4,5)P3 in membranes. As such, they are defining components of phosphoinositide-based cellular signaling and membrane trafficking pathways that regulate cell survival, growth, and proliferation, and are often deregulated in cancer. In this review, we highlight aspects of PI3Kα and PTEN interplay at the intersection of signaling and membrane trafficking. We also discuss the mechanisms of PI3Kα- and PTEN- membrane interaction and catalytic activation, which are fundamental for our understanding of the structural and allosteric implications on signaling at the membrane interface and may aid current efforts in pharmacological targeting of these proteins.
Collapse
Affiliation(s)
- Danai Maria Kotzampasi
- Biomedical Research Foundation, Academy of Athens, Athens 11527, Greece
- Department of Biology, University of Crete, Heraklion 71500, Greece
| | - Kyriaki Premeti
- Laboratory of Pharmacology, Faculty of Medicine, University of Ioannina, Ioannina 45110, Greece
| | - Alexandra Papafotika
- Laboratory of Biological Chemistry, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina 45110, Greece
- Biomedical Research Institute, Foundation for Research and Technology, Ioannina 45110, Greece
| | - Vasiliki Syropoulou
- Laboratory of Pharmacology, Faculty of Medicine, University of Ioannina, Ioannina 45110, Greece
| | - Savvas Christoforidis
- Laboratory of Biological Chemistry, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina 45110, Greece
- Biomedical Research Institute, Foundation for Research and Technology, Ioannina 45110, Greece
| | - Zoe Cournia
- Biomedical Research Foundation, Academy of Athens, Athens 11527, Greece
| | - George Leondaritis
- Laboratory of Pharmacology, Faculty of Medicine, University of Ioannina, Ioannina 45110, Greece
- Institute of Biosciences, University Research Center of Ioannina, Ioannina 45110, Greece
| |
Collapse
|
13
|
Li W, Zhang Z, Li Y, Wang Z. Abnormal hsa_circ_0003948 expression affects chronic periodontitis development by regulating miR-144-3p/NR2F2/PTEN signaling. J Periodontal Res 2021; 57:316-323. [PMID: 34910830 DOI: 10.1111/jre.12961] [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: 11/08/2021] [Accepted: 11/19/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND AND OBJECTIVE This study aimed to investigate the correlation between chronic periodontitis (CP) and abnormal circular RNA (circRNA) expression and to identify the role of hsa_circ_0003948 in the progression of CP. METHODS Next-generation sequencing was utilized to investigate abnormal expression of circRNA in gingival tissues from CP patients and healthy control subjects. Bioinformatics and luciferase reporting analyses were used to clarify the interactive relationship among circRNA, miRNA, and mRNA. Periodontal ligament cells (PDLCs) were employed to analyze proliferation and apoptosis after lipopolysaccharide (LPS) treatment using the cell counting kit 8 (CCK8) assay and flow cytometry detection. RESULTS High-throughput sequencing and RT-qPCR analyses confirmed that hsa_circ_0003948 expression decreased dramatically in gingival samples of CP patients. Overexpression of hsa_circ_0003948 alleviated LPS-induced PDLC injury by regulating NR2F2/PTEN signaling. The miR-144-3p and NR2F2 were determined to be hsa_circ_0003948 downstream targets. NR2F2 downregulation or miR-144-3p overexpression reversed the protective effect of hsa_circ_0003948 in PDLCs after treatment with LPS. Upregulation of NR2F2 reversed the inhibitory effect of miR-144-3p on surviving PDLCs after LPS treatment. CONCLUSION Overexpression of hsa_circ_0003948 exerts a protective effect in CP via miR-144-3p/NR2F2/PTEN signaling regulation.
Collapse
Affiliation(s)
- Wei Li
- Department of Stomatology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Zhi Zhang
- Department of Stomatology, Beijing Fengtai District Nanyuan Hospital, Beijing, China
| | - Yizi Li
- Department of Stomatology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Zuomin Wang
- Department of Stomatology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
14
|
Li Y, Fan C, Wang L, Lan T, Gao R, Wang W, Yu SY. MicroRNA-26a-3p rescues depression-like behaviors in male rats via preventing hippocampal neuronal anomalies. J Clin Invest 2021; 131:e148853. [PMID: 34228643 DOI: 10.1172/jci148853] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 07/01/2021] [Indexed: 12/11/2022] Open
Abstract
Depression is a neuropsychiatric disease associated with neuronal anomalies within specific brain regions. In the present study, we screened microRNA (miRNA) expression profiles in the dentate gyrus (DG) of the hippocampus and found that miR-26a-3p was markedly downregulated in a rat model of depression, whereas upregulation of miR-26a-3p within DG regions rescued the neuronal deterioration and depression-like phenotypes resulting from stress exposure, effects that appear to be mediated by the PTEN pathway. The knockdown of miR-26a-3p in DG regions of normal control rats induced depression-like behaviors, effects that were accompanied by activation of the PTEN/PI3K/Akt signaling pathway and neuronal deterioration via suppression of autophagy, impairments in synaptic plasticity, and promotion of neuronal apoptosis. In conclusion, these results suggest that miR-26a-3p deficits within the hippocampal DG mediated the neuronal anomalies contributing to the display of depression-like behaviors. This miRNA may serve as a potential therapeutic target for the treatment of depression.
Collapse
Affiliation(s)
- Ye Li
- Department of Physiology and
| | | | - Liyan Wang
- Morphological Experimental Center, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | | | - Rui Gao
- Department of Microorganism, Jinan Nursing Vocational College, Lvyoulu Road, Jinan, Shandong Province, China
| | | | - Shu Yan Yu
- Department of Physiology and.,Shandong Key Laboratory of Mental Disorders, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
15
|
Yu C, Xiong C, Tang J, Hou X, Liu N, Bayliss G, Zhuang S. Histone demethylase JMJD3 protects against renal fibrosis by suppressing TGFβ and Notch signaling and preserving PTEN expression. Am J Cancer Res 2021; 11:2706-2721. [PMID: 33456568 PMCID: PMC7806480 DOI: 10.7150/thno.48679] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 11/26/2020] [Indexed: 12/11/2022] Open
Abstract
Rationale: The Jumonji domain containing-3 (JMJD3), a specific histone demethylase for trimethylation on histone H3 lysine 27 (H3K27me3), is associated with the pathogenesis of many diseases, but its role in renal fibrosis remains unexplored. Here we examined the role of JMJD3 and mechanisms involved in the activation of renal fibroblasts and development of renal fibrosis. Methods: Murine models of 5/6 surgical nephrectomy (SNx) and ureteral unilateral obstruction (UUO) were used to assess the effect of a specific JMJD3 inhibitor, GSKJ4, and genetic deletion of JMJD3 from FOXD1 stroma-derived renal interstitial cells on the development of renal fibrosis and activation of renal interstitial fibroblasts. Cultured rat renal interstitial fibroblasts (NRK-49F) and mouse renal tubular epithelial cells (mTECs) were also used to examine JMJD3-mediated activation of profibrotic signaling. Results: JMJD3 and H3K27me3 expression levels were upregulated in the kidney of mice subjected to SNx 5/6 and UUO. Pharmacological inhibition of JMJD3 with GSKJ4 or genetic deletion of JMJD3 led to worsening of renal dysfunction as well as increased deposition of extracellular matrix proteins and activation of renal interstitial fibroblasts in the injured kidney. This was coincident with decreased expression of Smad7 and enhanced expression of H3K27me3, transforming growth factor β1 (TGFβ1), Smad3, Notch1, Notch3 and Jagged1. Inhibition of JMJD3 by GSK J4 or its specific siRNA also resulted in the similar responses in cultured NRK-49F and mTECs exposed to serum or TGFβ1. Moreover, JMJD3 inhibition augmented phosphorylation of AKT and ERK1/2 in vivo and in vitro. Conclusion: These results indicate that JMJD3 confers anti-fibrotic effects by limiting activation of multiple profibrotic signaling pathways and suggest that JMJD3 modulation may have therapeutic effects for chronic kidney disease.
Collapse
|