151
|
Ding L, Billadeau DD. Glycogen synthase kinase-3β: a novel therapeutic target for pancreatic cancer. Expert Opin Ther Targets 2020; 24:417-426. [PMID: 32178549 DOI: 10.1080/14728222.2020.1743681] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction: Pancreatic ductal adenocarcinoma (PDAC) is the third leading cause of cancer death in the United States with a single-digit 5-year survival rate despite advances in understanding the genetics and biology of the disease. Glycogen synthase kinase-3α (GSK-3α) and GSK-3β are serine/threonine kinases that localize to the cytoplasm, mitochondria and nucleus. Although they are highly homologous within their kinase domains and phosphorylate an overlapping set of target proteins, genetic studies have shown that GSK-3β regulates the activity of several proteins that promote neoplastic transformation. Significantly, GSK-3β is progressively overexpressed during PDAC development where it participates in tumor progression, survival and chemoresistance. Thus, GSK-3β has become an attractive target for treating PDAC.Areas covered: This review summarizes the mechanisms regulating GSK-3β activity, including upstream translational and post-translational regulation, as well as the downstream targets and their functions in PDAC cell growth, metastasis and chemoresistance.Expert opinion: The activity of GSK-3 kinases are considered cell- and context-specific. In PDAC, oncogenic KRas drives the transcriptional expression of the GSK-3β gene, which has been shown to regulate cancer cell proliferation and survival, as well as resistance to chemotherapy. Thus, the combination of GSK-3 inhibitors with chemotherapeutic drugs could be a promising strategy for PDAC.
Collapse
Affiliation(s)
- Li Ding
- The Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Daniel D Billadeau
- The Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
152
|
Nobakht N, Hanna RM, Al-Baghdadi M, Ameen KM, Arman F, Nobahkt E, Kamgar M, Rastogi A. Advances in Autosomal Dominant Polycystic Kidney Disease: A Clinical Review. Kidney Med 2020; 2:196-208. [PMID: 32734239 PMCID: PMC7380379 DOI: 10.1016/j.xkme.2019.11.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Polycystic kidney disease (PKD) is a multiorgan disorder resulting in fluid-filled cyst formation in the kidneys and other systems. The replacement of kidney parenchyma with an ever-increasing volume of cysts eventually leads to kidney failure. Recently, increased understanding of the pathophysiology of PKD and genetic advances have led to new approaches of treatment targeting physiologic pathways, which has been proven to slow the progression of certain types of the disease. We review the pathophysiologic patterns and recent advances in the clinical pharmacotherapy of autosomal dominant PKD. A multipronged approach with pharmacologic and nonpharmacologic treatments can be successfully used to slow down the rate of progression of autosomal dominant PKD to kidney failure.
Collapse
Affiliation(s)
- Niloofar Nobakht
- Division of Nephrology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Ramy M. Hanna
- Division of Nephrology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
- Division of Nephrology, Department of Medicine, University of California Irvine, Orange, CA
| | - Maha Al-Baghdadi
- Division of Nephrology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
- Department of Medicine, University of Alabama Birmingham Huntsville Regional Campus, Huntsville, AL
| | - Khalid Mohammed Ameen
- Division of Nephrology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Farid Arman
- Division of Nephrology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
- Department of Medicine, University of Pennsylvania Medical Center, Philadelphia, PA
| | - Ehsan Nobahkt
- Division of Renal Diseases and Hypertension, Department of Medicine, George Washington University, Washington, DC
| | - Mohammad Kamgar
- Division of Nephrology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Anjay Rastogi
- Division of Nephrology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| |
Collapse
|
153
|
Sulfhydration of AKT triggers Tau-phosphorylation by activating glycogen synthase kinase 3β in Alzheimer's disease. Proc Natl Acad Sci U S A 2020; 117:4418-4427. [PMID: 32051249 DOI: 10.1073/pnas.1916895117] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In Alzheimer's disease (AD), human Tau is phosphorylated at S199 (hTau-S199-P) by the protein kinase glycogen synthase kinase 3β (GSK3β). HTau-S199-P mislocalizes to dendritic spines, which induces synaptic dysfunction at the early stage of AD. The AKT kinase, once phosphorylated, inhibits GSK3β by phosphorylating it at S9. In AD patients, the abundance of phosphorylated AKT with active GSK3β implies that phosphorylated AKT was unable to inactivate GSK3β. However, the underlying mechanism of the inability of phosphorylated AKT to phosphorylate GSK3β remains unknown. Here, we show that total AKT and phosphorylated AKT was sulfhydrated at C77 due to the induction of intracellular hydrogen sulfide (H2S). The increase in intracellular H2S levels resulted from the induction of the proinflammatory cytokine, IL-1β, which is a pathological hallmark of AD. Sulfhydrated AKT does not interact with GSK3β, and therefore does not phosphorylate GSK3β. Thus, active GSK3β phosphorylates Tau aberrantly. In a transgenic knockin mouse (AKT-KI+/+) that lacked sulfhydrated AKT, the interaction between AKT or phospho-AKT with GSK3β was restored, and GSK3β became phosphorylated. In AKT-KI+/+ mice, expressing the pathogenic human Tau mutant (hTau-P301L), the hTau S199 phosphorylation was ameliorated as GSK3β phosphorylation was regained. This event leads to a decrease in dendritic spine loss by reducing dendritic localization of hTau-S199-P, which improves cognitive dysfunctions. Sulfhydration of AKT was detected in the postmortem brains from AD patients; thus, it represents a posttranslational modification of AKT, which primarily contributes to synaptic dysfunction in AD.
Collapse
|
154
|
Kim JW, Yang JH, Kim EJ. SIRT1 and AROS suppress doxorubicin-induced apoptosis via inhibition of GSK3β activity in neuroblastoma cells. Anim Cells Syst (Seoul) 2020; 24:53-59. [PMID: 32158616 PMCID: PMC7048222 DOI: 10.1080/19768354.2020.1726461] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/14/2020] [Accepted: 02/03/2020] [Indexed: 01/27/2023] Open
Abstract
SIRT1, the best-characterized member of the sirtuin family of deacetylases, is involved in cancer, apoptosis, inflammation, and metabolism. Active regulator of SIRT1 (AROS) was the first identified direct regulator of SIRT1. An increasing number of reports have indicated that SIRT1 plays an important role in controlling brain tumors. Here, we demonstrated that depletion of SIRT1 and AROS increases doxorubicin-mediated apoptosis in human neuroblastoma SH-SY5Y cells. Glycogen synthase kinase 3β (GSK3β) promoted doxorubicin-mediated apoptosis, but this effect was abolished by overexpression of SIRT1 and AROS. Interestingly, SIRT1 and AROS interacted with GSK3β and increased inhibitory phosphorylation of GSK3β on Ser9. Finally, we determined that AROS cooperates with SIRT1 to suppress GSK3β acetylation. Taken together, our results suggest that SIRT1 and AROS inhibit GSK3β activity and provide additional insight into drug resistance in the treatment of neuroblastoma.
Collapse
Affiliation(s)
- Jeong Woo Kim
- Department of Molecular Biology, Dankook University, Cheonan-si, Korea
| | - Ji Hye Yang
- Department of Molecular Biology, Dankook University, Cheonan-si, Korea
| | - Eun-Joo Kim
- Department of Molecular Biology, Dankook University, Cheonan-si, Korea
| |
Collapse
|
155
|
Alcaraz E, Vilardell J, Borgo C, Sarró E, Plana M, Marin O, Pinna LA, Bayascas JR, Meseguer A, Salvi M, Itarte E, Ruzzene M. Effects of CK2β subunit down-regulation on Akt signalling in HK-2 renal cells. PLoS One 2020; 15:e0227340. [PMID: 31910234 PMCID: PMC6946142 DOI: 10.1371/journal.pone.0227340] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 12/17/2019] [Indexed: 12/25/2022] Open
Abstract
The PI3K/Akt pathway is interconnected to protein kinase CK2, which directly phosphorylates Akt1 at S129. We have previously found that, in HK-2 renal cells, downregulation of the CK2 regulatory subunit β (shCK2β cells) reduces S129 Akt phosphorylation. Here, we investigated in more details how the different CK2 isoforms impact on Akt and other signaling pathways. We found that all CK2 isoforms phosphorylate S129 in vitro, independently of CK2β. However, in HK-2 cells the dependence on CK2β was confirmed by rescue experiments (CK2β re-expression in shCK2β HK-2 cells), suggesting the presence of additional components that drive Akt recognition by CK2 in cells. We also found that CK2β downregulation altered the phosphorylation ratio between the two canonical Akt activation sites (pT308 strongly reduced, pS473 slightly increased) in HK-2 cells. Similar results were found in other cell lines where CK2β was stably knocked out by CRISPR-Cas9 technology. The phosphorylation of rpS6 S235/S236, a downstream effector of Akt, was strongly reduced in shCK2β HK-2 cells, while the phosphorylation of two Akt direct targets, PRAS40 T246 and GSK3β S9, was increased. Differently to what observed in response to CK2β down-regulation, the chemical inhibition of CK2 activity by cell treatment with the specific inhibitor CX-4945 reduced both the Akt canonical sites, pT308 and pS473. In CX-4945-treated cells, the changes in rpS6 pS235/S236 and GSK3β pS9 mirrored those induced by CK2β knock-down (reduction and slight increase, respectively); on the contrary, the effect on PRAS40 pT246 phosphorylation was sharply different, being strongly reduced by CK2 inhibition; this suggests that this Akt target might be dependent on Akt pS473 status in HK-2 cells. Since PI3K/Akt and ERK1/2/p90rsk pathways are known to be interconnected and both modulated by CK2, with GSK3β pS9 representing a convergent point, we investigated if ERK1/2/p90rsk signaling was affected by CK2β knock-down and CX-4945 treatment in HK-2 cells. We found that p90rsk was insensitive to any kind of CK2 targeting; therefore, the observation that, similarly, GSK3β pS9 was not reduced by CK2 blockade suggests that GSK3β phosphorylation is mainly under the control of p90rsk in these cells. However, we found that the PI3K inhibitor LY294002 reduced GSK3β pS9, and concomitantly decreased Snail1 levels (a GSK3β target and Epithelial-to-Mesenchymal transition marker). The effects of LY294002 were observed also in CK2β-downregulated cells, suggesting that reducing GSK3β pS9 could be a strategy to control Snail1 levels in any situation where CK2β is defective, as possibly occurring in cancer cells.
Collapse
Affiliation(s)
- Estefania Alcaraz
- Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra (Barcelona) Spain
| | - Jordi Vilardell
- Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra (Barcelona) Spain
| | - Christian Borgo
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Eduard Sarró
- Fisiopatología Renal, CIBBIM-Nanomedicine, VHIR, Barcelona, Spain
| | - Maria Plana
- Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra (Barcelona) Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Cerdanyola del Vallès, Barcelona, Spain
| | - Oriano Marin
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Lorenzo A. Pinna
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- CNR Neuroscience Institute, Padova, Italy
| | - José R. Bayascas
- Departament de Bioquimica i Biologia Molecular, Unitat de Bioquímica de Medicina, Universitat Autònoma de Barcelona, Bellaterra (Barcelona) Spain
- Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Anna Meseguer
- Fisiopatología Renal, CIBBIM-Nanomedicine, VHIR, Barcelona, Spain
- Departament de Bioquimica i Biologia Molecular, Unitat de Bioquímica de Medicina, Universitat Autònoma de Barcelona, Bellaterra (Barcelona) Spain
- Red de Investigación Renal (REDINREN), Instituto de Salud Carlos III-FEDER, Madrid, Spain
| | - Mauro Salvi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Emilio Itarte
- Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra (Barcelona) Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Cerdanyola del Vallès, Barcelona, Spain
| | - Maria Ruzzene
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- CNR Neuroscience Institute, Padova, Italy
| |
Collapse
|
156
|
Chang PK, Chu J, Tsai YT, Lai YH, Chen JC. Dopamine D 3 receptor and GSK3β signaling mediate deficits in novel object recognition memory within dopamine transporter knockdown mice. J Biomed Sci 2020; 27:16. [PMID: 31900153 PMCID: PMC6942274 DOI: 10.1186/s12929-019-0613-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 12/29/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Over-stimulation of dopamine signaling is thought to underlie the pathophysiology of a list of mental disorders, such as psychosis, mania and attention-deficit/hyperactivity disorder. These disorders are frequently associated with cognitive deficits in attention or learning and memory, suggesting that persistent activation of dopamine signaling may change neural plasticity to induce cognitive or emotional malfunction. METHODS Dopamine transporter knockdown (DAT-KD) mice were used to mimic a hyper-dopamine state. Novel object recognition (NOR) task was performed to assess the recognition memory. To test the role of dopamine D3 receptor (D3R) on NOR, DAT-KD mice were treated with either a D3R antagonist, FAUC365 or by deletion of D3R. Total or phospho-GSK3 and -ERK1/2 signals in various brain regions were measured by Western blot analyses. To examine the impact of GSK3 signal on NOR, wild-type mice were systemically treated with GSK3 inhibitor SB216763 or, micro-injected with lentiviral shRNA of GSK3β or GSK3α in the medial prefrontal cortex (mPFC). RESULTS We confirmed our previous findings that DAT-KD mice displayed a deficit in NOR memory, which could be prevented by deletion of D3R or exposure to FAUC365. In WT mice, p-GSK3α and p-GSK3β were significantly decreased in the mPFC after exposure to novel objects; however, the DAT-KD mice exhibited no such change in mPFC p-GSK3α/β levels. DAT-KD mice treated with FAUC365 or with D3R deletion exhibited restored novelty-induced GSK3 dephosphorylation in the mPFC. Moreover, inhibition of GSK3 in WT mice diminished NOR performance and impaired recognition memory. Lentiviral shRNA knockdown of GSK3β, but not GSK3α, in the mPFC of WT mice also impaired NOR. CONCLUSION These findings suggest that D3R acts via GSK3β signaling in the mPFC to play a functional role in NOR memory. In addition, treatment with D3R antagonists may be a reasonable approach for ameliorating cognitive impairments or episodic memory deficits in bipolar disorder patients.
Collapse
Affiliation(s)
- Pi-Kai Chang
- Department of Physiology and Pharmacology, Graduate Institute of Biomedical Sciences, School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Jung Chu
- Department of Biomedical Sciences, School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ya-Ting Tsai
- Department of Biomedical Sciences, School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yan-Heng Lai
- Department of Medical Imaging and Radiological Sciences, School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Jin-Chung Chen
- Department of Physiology and Pharmacology, Graduate Institute of Biomedical Sciences, School of Medicine, Chang Gung University, Taoyuan, Taiwan. .,Healthy Ageing Research Center, Chang Gung University, Taoyuan, Taiwan. .,Neuroscience Research Center, Chang Gung Memorial Hospital, Linko, Taiwan.
| |
Collapse
|
157
|
Evangelisti C, Chiarini F, Paganelli F, Marmiroli S, Martelli AM. Crosstalks of GSK3 signaling with the mTOR network and effects on targeted therapy of cancer. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1867:118635. [PMID: 31884070 DOI: 10.1016/j.bbamcr.2019.118635] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 12/18/2019] [Indexed: 02/06/2023]
Abstract
The introduction of therapeutics targeting specific tumor-promoting oncogenic or non-oncogenic signaling pathways has revolutionized cancer treatment. Mechanistic (previously mammalian) target of rapamycin (mTOR), a highly conserved Ser/Thr kinase, is a central hub of the phosphatidylinositol 3-kinase (PI3K)/Akt/mTOR network, one of the most frequently deregulated signaling pathways in cancer, that makes it an attractive target for therapy. Numerous mTOR inhibitors have progressed to clinical trials and two of them have been officially approved as anticancer therapeutics. However, mTOR-targeting drugs have met with a very limited success in cancer patients. Frequently, the primary impediment to a successful targeted therapy in cancer is drug-resistance, either from the very beginning of the therapy (innate resistance) or after an initial response and upon repeated drug treatment (evasive or acquired resistance). Drug-resistance leads to treatment failure and relapse/progression of the disease. Resistance to mTOR inhibitors depends, among other reasons, on activation/deactivation of several signaling pathways, included those regulated by glycogen synthase kinase-3 (GSK3), a protein that targets a vast number of substrates in its repertoire, thereby orchestrating many processes that include cell proliferation and survival, metabolism, differentiation, and stemness. A detailed knowledge of the rewiring of signaling pathways triggered by exposure to mTOR inhibitors is critical to our understanding of the consequences such perturbations cause in tumors, including the emergence of drug-resistant cells. Here, we provide the reader with an updated overview of intricate circuitries that connect mTOR and GSK3 and we relate them to the efficacy (or lack of efficacy) of mTOR inhibitors in cancer cells.
Collapse
Affiliation(s)
- Camilla Evangelisti
- CNR Institute of Molecular Genetics, 40136 Bologna, BO, Italy; IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, BO, Italy
| | - Francesca Chiarini
- CNR Institute of Molecular Genetics, 40136 Bologna, BO, Italy; IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, BO, Italy
| | - Francesca Paganelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, BO, Italy
| | - Sandra Marmiroli
- Department of Biomedical, Metabolical, and Neurological Sciences, University of Modena and Reggio Emilia, 41124 Modena, MO, Italy
| | - Alberto M Martelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, BO, Italy.
| |
Collapse
|
158
|
Gong Y, Zhang Z, Chang Z, Zhou H, Zhao R, He B. Inactivation of glycogen synthase kinase-3α is required for mitochondria-mediated apoptotic germ cell phagocytosis in Sertoli cells. Aging (Albany NY) 2019; 10:3104-3116. [PMID: 30398976 PMCID: PMC6286816 DOI: 10.18632/aging.101614] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 10/19/2018] [Indexed: 12/15/2022]
Abstract
The rapid and efficient clearance of apoptotic germ cells (GCs) by Sertoli cells (SCs) is important for spermatogenesis. High mitochondrial activity in phagocytes is critical for continued clearance of apoptotic cells. However, the underlying molecular mechanism is poorly understood. Glycogen synthase kinase-3α (GSK3α) is a protein kinase that participates in the regulation of mitochondrial activity. Immunohistochemistry evidenced the predominant presence of the Ser21 phosphorylation GSK3α (inactivation) signal in SCs. Heat shock-induced apoptosis of GCs and dephosphorylation of GSK3α in SCs is a perfect model to investigate the role of GSK3α in phagocytic action. The number of apoptotic GCs was significantly lower in GSK3α inhibitor pre-treated mice with HS compared to normal control. In vitro phagocytosis assays shown that the phagocytic activity in GSK3α activated SCs was downregulated, while GSK3α inhibitor supplementation restored this process. Moreover, GSK3α activation participates in the alteration of the mitochondrial ultrastructure and activity. In particular, GSK3α activation inhibits mitochondrial fission via phosphorylation of dynamin related protein 1 at Ser637. Changes of mitochondrial activity resulted in the accumulation of lipid droplets and the alteration of metabolism pattern in SCs. In summary, our results demonstrate that inactivation of GSK3α is required for mitochondria-mediated apoptotic GCs phagocytosis in SCs.
Collapse
Affiliation(s)
- Yabin Gong
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Zhilong Zhang
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Zhanglin Chang
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Hao Zhou
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Ruqian Zhao
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing Agricultural University, Nanjing 210095, PR China.,Jiangsu Collaborative Innovation Centre of Meat Production and Processing, Quality and Safety Control, Nanjing 210095, PR China
| | - Bin He
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing Agricultural University, Nanjing 210095, PR China.,Jiangsu Collaborative Innovation Centre of Meat Production and Processing, Quality and Safety Control, Nanjing 210095, PR China
| |
Collapse
|
159
|
Dey S, Brothag C, Vijayaraghavan S. Signaling Enzymes Required for Sperm Maturation and Fertilization in Mammals. Front Cell Dev Biol 2019; 7:341. [PMID: 31921853 PMCID: PMC6930163 DOI: 10.3389/fcell.2019.00341] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 12/03/2019] [Indexed: 12/18/2022] Open
Abstract
In mammals, motility and fertilizing ability of spermatozoa develop during their passage through the epididymis. After ejaculation, sperm undergo capacitation and hyperactivation in the female reproductive tract - a motility transition that is required for sperm penetration of the egg. Both epididymal initiation of sperm motility and hyperactivation are essential for male fertility. Motility initiation in the epididymis and sperm hyperactivation involve changes in metabolism, cAMP (cyclic adenosine mono-phosphate), calcium and pH acting through protein kinases and phosphatases. Despite this knowledge, we still do not understand, in biochemical terms, how sperm acquire motility in the epididymis and how motility is altered in the female reproductive tract. Recent data show that the sperm specific protein phosphatase PP1γ2, glycogen synthase kinase 3 (GSK3), and the calcium regulated phosphatase calcineurin (PP2B), are involved in epididymal sperm maturation. The protein phosphatase PP1γ2 is present only in testis and sperm in mammals. PP1γ2 has a isoform-specific requirement for normal function of mammalian sperm. Sperm PP1γ2 is regulated by three proteins - inhibitor 2, inhibitor 3 and SDS22. Changes in phosphorylation of these three inhibitors and their binding to PP1γ2 are involved in initiation and activation of sperm motility. The inhibitors are phosphorylated by protein kinases, one of which is GSK3. The isoform GSK3α is essential for epididymal sperm maturation and fertility. Calcium levels dramatically decrease during sperm maturation and initiation of motility suggesting that the calcium activated sperm phosphatase (PP2B) activity also decreases. Loss of PP2B results in male infertility due to impaired sperm maturation in the epididymis. Thus the three signaling enzymes PP1γ2, GSK3, and PP2B along with the documented PKA (protein kinase A) have key roles in sperm maturation and hyperactivation. Significantly, all these four signaling enzymes are present as specific isoforms only in placental mammals, a testimony to their essential roles in the unique aspects of sperm function in mammals. These findings should lead to a better biochemical understanding of the basis of male infertility and should lead to novel approaches to a male contraception and managed reproduction.
Collapse
|
160
|
GSK3β Regulates Brain Energy Metabolism. Cell Rep 2019; 23:1922-1931.e4. [PMID: 29768193 PMCID: PMC6082412 DOI: 10.1016/j.celrep.2018.04.045] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 01/08/2018] [Accepted: 04/11/2018] [Indexed: 01/01/2023] Open
Abstract
GSK3β is a serine threonine kinase implicated in the progression of Alzheimer’s disease. Although the role of GSK3β in growth and pathology has been extensively studied, little is known about the metabolic consequences of GSK3β manipulation, particularly in the brain. Here, we show that GSK3β regulates mitochondrial energy metabolism in human H4 neuroglioma cells and rat PC12-derived neuronal cells and that inhibition of GSK3β in mice in vivo alters metabolism in the hippocampus in a region-specific manner. We demonstrate that GSK3β inhibition increases mitochondrial respiration and membrane potential and alters NAD(P)H metabolism. These metabolic effects are associated with increased PGC-1α protein stabilization, enhanced nuclear localization, and increased transcriptional co-activation. In mice treated with the GSK3β inhibitor lithium carbonate, changes in hippocampal energy metabolism are linked to increased PGC-1α. These data highlight a metabolic role for brain GSK3β and suggest that the GSK3β/PGC-1α axis may be important in neuronal metabolic integrity.
Collapse
|
161
|
Piazza F, Manni S, Arjomand A, Visentin A, Trentin L, Semenzato G. New responsibilities for aged kinases in B-lymphomas. Hematol Oncol 2019; 38:3-11. [PMID: 31782972 DOI: 10.1002/hon.2694] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 11/05/2019] [Indexed: 12/21/2022]
Abstract
The knowledge accumulated over the last decade on B-cell-derived non-Hodgkin lymphoma (B-NHL) pathogenesis has led to the identification of several molecular abnormalities, opening new perspectives in the design of novel therapies. Indeed, drugs targeting specific biochemical pathways critical for B-NHL cell survival, proliferation, and fitness within the malignant microenvironment are now available to the clinician: the B-cell receptor signaling inhibitors of BTK, PI3Kδ, ζ, γ, and SYK or the pro-apoptotic BH3-mimetics are clear examples of it. Moreover, it is emerging that malignant B-cell growth is sustained not only by mutations in oncogenes/tumor suppressors but also by the "addiction" to nononcogene (ie, nonstructurally altered) molecules. In this regard, a consistent body of data has established that the Ser/Thr kinases CK1, CK2, and GSK3 are involved in malignant lymphocyte biology and act as pro-survival and signaling-boosting molecules, both in precursor and mature B-cell tumors. Currently, an experimental and clinical groundwork is available, upon which to design CK1-, CK2-, and GSK3-directed antilymphoma/leukemia therapies. In this review, we have examined the main features of CK1, CK2, and GSK3 kinases, summarized the data in B-NHL supporting them as suitable therapeutic targets, and proposed a perspective on potential future research development.
Collapse
Affiliation(s)
- Francesco Piazza
- Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,Unit of Hematological Malignancies - Laboratory of Myeloma and Lymphoma Pathobiology, Foundation for Advanced Biomedical Research - Veneto Institute of Molecular Medicine (FABR-VIMM), Padova, Italy
| | - Sabrina Manni
- Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,Unit of Hematological Malignancies - Laboratory of Myeloma and Lymphoma Pathobiology, Foundation for Advanced Biomedical Research - Veneto Institute of Molecular Medicine (FABR-VIMM), Padova, Italy
| | - Arash Arjomand
- Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,Unit of Hematological Malignancies - Laboratory of Myeloma and Lymphoma Pathobiology, Foundation for Advanced Biomedical Research - Veneto Institute of Molecular Medicine (FABR-VIMM), Padova, Italy
| | - Andrea Visentin
- Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,Unit of Hematological Malignancies - Laboratory of Myeloma and Lymphoma Pathobiology, Foundation for Advanced Biomedical Research - Veneto Institute of Molecular Medicine (FABR-VIMM), Padova, Italy
| | - Livio Trentin
- Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,Unit of Hematological Malignancies - Laboratory of Myeloma and Lymphoma Pathobiology, Foundation for Advanced Biomedical Research - Veneto Institute of Molecular Medicine (FABR-VIMM), Padova, Italy
| | - Gianpietro Semenzato
- Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,Unit of Hematological Malignancies - Laboratory of Myeloma and Lymphoma Pathobiology, Foundation for Advanced Biomedical Research - Veneto Institute of Molecular Medicine (FABR-VIMM), Padova, Italy
| |
Collapse
|
162
|
Involvement of PI3K/Akt/GSK-3β signaling pathway in the antidepressant-like and neuroprotective effects of Morus nigra and its major phenolic, syringic acid. Chem Biol Interact 2019; 314:108843. [DOI: 10.1016/j.cbi.2019.108843] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 08/11/2019] [Accepted: 10/02/2019] [Indexed: 01/31/2023]
|
163
|
Alhusaini A, Fadda L, Hasan IH, Ali HM, El Orabi NF, Badr AM, Zakaria E, Alenazi AM, Mahmoud AM. Arctium lappa Root Extract Prevents Lead-Induced Liver Injury by Attenuating Oxidative Stress and Inflammation, and Activating Akt/GSK-3β Signaling. Antioxidants (Basel) 2019; 8:582. [PMID: 31771282 PMCID: PMC6943639 DOI: 10.3390/antiox8120582] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 11/15/2019] [Accepted: 11/22/2019] [Indexed: 12/26/2022] Open
Abstract
Arctium lappa L (A. lappa) is a popular medicinal plant with promising hepatoprotective activity. This study investigated the protective effect of A. lappa root extract (ALRE) on lead (Pb) hepatotoxicity, pointing to its ability to modulate oxidative stress, inflammation, and protein kinase B/Akt/glycogen synthase kinase (GSK)-3β signaling. Rats received 50 mg/kg lead acetate (Pb(Ac)2) and 200 mg/kg ALRE or vitamin C (Vit. C) for 7 days, and blood and liver samples were collected. Pb(Ac)2 provoked hepatotoxicity manifested by elevated serum transaminases and lactate dehydrogenase, and decreased total protein. Histopathological alterations, including distorted lobular hepatic architecture, microsteatotic changes, congestion, and massive necrosis were observed in Pb(II)-induced rats. ALRE ameliorated liver function and prevented all histological alterations. Pb(II) increased hepatic lipid peroxidation (LPO), nitric oxide (NO), caspase-3, and DNA fragmentation, and serum C-reactive protein, tumor necrosis factor-α, and interleukin-1β. Cellular antioxidants, and Akt and GSK-3β phosphorylation levels were decreased in the liver of Pb(II)-induced rats. ALRE ameliorated LPO, NO, caspase-3, DNA fragmentation and inflammatory mediators, and boosted antioxidant defenses in Pb(II)-induced rats. In addition, ALRE activated Akt and inhibited GSK-3β in the liver of Pb(II)-induced rats. In conclusion, ALRE inhibits liver injury in Pb(II)-intoxicated rats by attenuating oxidative injury and inflammation, and activation of Akt/GSK-3β signaling pathway.
Collapse
Affiliation(s)
- Ahlam Alhusaini
- Pharmacology and Toxicology Department, Faculty of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (L.F.); (I.H.H.); (N.F.E.O.); (A.M.B.); (A.M.A.)
| | - Laila Fadda
- Pharmacology and Toxicology Department, Faculty of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (L.F.); (I.H.H.); (N.F.E.O.); (A.M.B.); (A.M.A.)
| | - Iman H. Hasan
- Pharmacology and Toxicology Department, Faculty of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (L.F.); (I.H.H.); (N.F.E.O.); (A.M.B.); (A.M.A.)
| | - Hanaa M. Ali
- Common First Year Deanship, King Saud University, Riyadh 11451, Saudi Arabia;
- Genetic and Cytology Department, National Research Centre, Giza 12622, Egypt
| | - Naglaa F. El Orabi
- Pharmacology and Toxicology Department, Faculty of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (L.F.); (I.H.H.); (N.F.E.O.); (A.M.B.); (A.M.A.)
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Amira M. Badr
- Pharmacology and Toxicology Department, Faculty of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (L.F.); (I.H.H.); (N.F.E.O.); (A.M.B.); (A.M.A.)
- Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Enas Zakaria
- Pharmaceutics Department, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Abeer M. Alenazi
- Pharmacology and Toxicology Department, Faculty of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (L.F.); (I.H.H.); (N.F.E.O.); (A.M.B.); (A.M.A.)
| | - Ayman M. Mahmoud
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef 62514, Egypt
| |
Collapse
|
164
|
Alhusaini A, Fadda L, Hasan IH, Zakaria E, Alenazi AM, Mahmoud AM. Curcumin Ameliorates Lead-Induced Hepatotoxicity by Suppressing Oxidative Stress and Inflammation, and Modulating Akt/GSK-3β Signaling Pathway. Biomolecules 2019; 9:biom9110703. [PMID: 31694300 PMCID: PMC6920970 DOI: 10.3390/biom9110703] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 10/15/2019] [Accepted: 11/04/2019] [Indexed: 12/14/2022] Open
Abstract
Lead (Pb) is a toxic heavy metal pollutant with adverse effects on the liver and other body organs. Curcumin (CUR) is the principal curcuminoid of turmeric and possesses strong antioxidant and anti-inflammatory activities. This study explored the protective effect of CUR on Pb hepatotoxicity with an emphasis on oxidative stress, inflammation and Akt/GSK-3β signaling. Rats received lead acetate and CUR and/or ascorbic acid (AA) for seven days and samples were collected for analyses. Pb(II) induced liver injury manifested by elevated serum alanine aminotransferase (ALT), aspartate aminotransferase (AST) and lactate dehydrogenase (LDH), as well as histopathological alterations, including massive hepatocyte degeneration and increased collagen deposition. Lipid peroxidation, nitric oxide, TNF-α and DNA fragmentation were increased, whereas antioxidant defenses were diminished in the liver of Pb(II)-intoxicated rats. Pb(II) increased hepatic NF-κB and JNK phosphorylation and caspase-3 cleavage, whereas Akt and GSK-3β phosphorylation was decreased. CUR and/or AA ameliorated liver function, prevented tissue injury, and suppressed oxidative stress, DNA damage, NF-κB, JNK and caspase-3. In addition, CUR and/or AA activated Akt and inhibited GSK-3β in Pb(II)-induced rats. In conclusion, CUR prevents Pb(II) hepatotoxicity via attenuation of oxidative injury and inflammation, activation of Akt and inhibition of GSK-3β. However, further studies scrutinizing the exact role of Akt/GSK-3β signaling are recommended.
Collapse
Affiliation(s)
- Ahlam Alhusaini
- Pharmacology and Toxicology Department, Faculty of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (L.F.); (I.H.H.); (A.M.A.)
- Correspondence: (A.A.); (A.M.M.)
| | - Laila Fadda
- Pharmacology and Toxicology Department, Faculty of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (L.F.); (I.H.H.); (A.M.A.)
| | - Iman H. Hasan
- Pharmacology and Toxicology Department, Faculty of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (L.F.); (I.H.H.); (A.M.A.)
| | - Enas Zakaria
- Pharmaceutics Department, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Abeer M. Alenazi
- Pharmacology and Toxicology Department, Faculty of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (L.F.); (I.H.H.); (A.M.A.)
| | - Ayman M. Mahmoud
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef 62514, Egypt
- Correspondence: (A.A.); (A.M.M.)
| |
Collapse
|
165
|
Wang L, Liu X, Zhan S, Guo J, Yang S, Zhong T, Li L, Zhang H, Wang Y. Inhibition of GSK3β Reduces Ectopic Lipid Accumulation and Induces Autophagy by the AMPK Pathway in Goat Muscle Satellite Cells. Cells 2019; 8:cells8111378. [PMID: 31683987 PMCID: PMC6912237 DOI: 10.3390/cells8111378] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 10/27/2019] [Accepted: 10/31/2019] [Indexed: 12/16/2022] Open
Abstract
Ectopic lipid accumulation in muscle is important not only for obesity and myopathy treatment, but also for meat quality improvement in farm animals. However, the molecular mechanisms involved in lipid metabolism in muscle satellite cells are still elusive. In this study, SB216763 reduced GSK3β activation by increasing the level of pGSK3β (Ser9) and decreasing the level of total GSK3β protein. GSK3β inhibition decreased lipid accumulation and downregulated the expression level of lipogenesis-related genes in the adipogenic differentiation of goat muscle satellite cells. Furthermore, SB216763 treatment increased the levels of pAMPKα (T172) and pACC (Ser79). Further, we found that GSK3β inhibition promoted levels of LC3B-II and reduced the protein levels of p62 to induce the autophagy in muscle satellite cells. Taken together, our results provide new insight into a critical function for GSK3β: modulating lipid accumulation in goat muscle satellite cells through activating the AMPK pathway.
Collapse
Affiliation(s)
- Linjie Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| | - Xin Liu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| | - Siyuan Zhan
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| | - Jiazhong Guo
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| | - Shizhong Yang
- Institute of Liangshan Animal Husbandry and Veterinary Science, Xichang 615042, Sichuan, China.
| | - Tao Zhong
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| | - Li Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| | - Hongping Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| | - Yan Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| |
Collapse
|
166
|
Bhattacharjee R, Goswami S, Dey S, Gangoda M, Brothag C, Eisa A, Woodgett J, Phiel C, Kline D, Vijayaraghavan S. Isoform-specific requirement for GSK3α in sperm for male fertility. Biol Reprod 2019; 99:384-394. [PMID: 29385396 DOI: 10.1093/biolre/ioy020] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 01/22/2018] [Indexed: 12/28/2022] Open
Abstract
Glycogen synthase kinase 3 (GSK3) is a highly conserved protein kinase regulating key cellular functions. Its two isoforms, GSK3α and GSK3β, are encoded by distinct genes. In most tissues the two isoforms are functionally interchangeable, except in the developing embryo where GSK3β is essential. One functional allele of either of the two isoforms is sufficient to maintain normal tissue functions. Both GSK3 isoforms, present in sperm from several species including human, are suggested to play a role in epididymal initiation of sperm motility. Using genetic approaches, we have tested requirement for each of the two GSK3 isoforms in testis and sperm. Both GSK3 isoforms are expressed at high levels during the onset of spermatogenesis. Conditional knockout of GSK3α, but not GSK3β, in developing testicular germ cells in mice results in male infertility. Mice lacking one allele each of GSK3α and GSK3β are fertile. Despite overlapping expression and localization in differentiating spermatids, GSK3β does not substitute for GSK3α. Loss of GSK3α impairs sperm hexokinase activity resulting in low ATP levels. Net adenine nucleotide levels in caudal sperm lacking GSK3α resemble immature caput epididymal sperm. Changes in the association of the protein phosphatase PP1γ2 with its protein interactors occurring during epididymal sperm maturation is impaired in sperm lacking GSK3α. The isoform-specific requirement for GSK3α is likely due to its specific binding partners in the sperm principal piece. Testis and sperm are unique in their specific requirement of GSK3α for normal function and male fertility.
Collapse
Affiliation(s)
| | - Suranjana Goswami
- Department of Biological Sciences, Kent State University, Kent, Ohio, USA
| | - Souvik Dey
- Department of Biological Sciences, Kent State University, Kent, Ohio, USA
| | - Mahinda Gangoda
- Department of Chemistry and Biochemistry, Kent State University, Kent, Ohio, USA
| | - Cameron Brothag
- School of Biomedical Sciences, Kent State University, Kent, Ohio, USA
| | - Alaa Eisa
- School of Biomedical Sciences, Kent State University, Kent, Ohio, USA
| | - James Woodgett
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Christopher Phiel
- Department of Integrative Biology, University of Colorado Denver, Denver, Colorado, USA
| | - Douglas Kline
- Department of Biological Sciences, Kent State University, Kent, Ohio, USA
| | | |
Collapse
|
167
|
Xu K, Xie X, Qi G, Weng P, Hu Z, Han K, Yuan H, Li Y, Ran X, Lin G, Hu C. Grass carp STK38 regulates IFN I expression by decreasing the phosphorylation level of GSK3β. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 99:103410. [PMID: 31175887 DOI: 10.1016/j.dci.2019.103410] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 06/04/2019] [Accepted: 06/04/2019] [Indexed: 06/09/2023]
Abstract
As a member of NDR protein kinase family and a novel protein kinase of Hippo signal pathway, Serine/threonine kinase 38 (STK38) plays a very significant role in the innate immune. In mammals, STK38 performs its function by combining with GSK3β. Nowadays, there are few reports of STK38 in fish. In order to explain the function of fish STK38 in the innate immunity, we cloned the ORF of grass carp (Ctenopharyngodon idella) STK38 (CiSTK38) and the related kinase GSK3β (CiGSK3β). Phylogenetic trees revealed that CiSTK38 and CiGSK3β evolved closer kinship with sinocyclocheilus grahami STK38 and siniperca chuatsi GSK3β respectively. CiSTK38 and CiGSK3β can respond to the intradermal injection of poly (I:C) in grass carp different tissues and the transfection of poly I:C in CIK cells. Subcellular localization revealed the CiGSK3β were broadly distributed through the cytoplasm, whereas CiSTK38 were observed both in cytoplasm and nucleus. However, when they were co-transferred into cells, the two proteins were found to aggregate in the nucleus. GST-pulldown and co-immunoprecipitation analysis revealed that CiSTK38 can physically interact with CiGSK3β. Phos-tag PAGE illustrated CiSTK38 can decrease the phosphorylation and auto-phosphorylation level of CiGSK3β at Ser9 and at Tyr216. To investigate the functional correlation between CiSTK38 and CiGSK3β, we overexpressed CiSTK38 and CiGSK3β in CIK cells and found that they can up-regulate the expression of IFN I. In short, we demonstrated that CiSTK38 can confer CiGSK3β kinase activity by reducing its phosphorylation level. Result from this study strongly suggested that the anti-viral immune effects elicited by poly (I:C) in part were mediated through activation of CiGSK3β. The findings provided scientific basis for the anti-viral immune mechanism of STK38 and GSK3β in fish.
Collapse
Affiliation(s)
- Kang Xu
- College of Life Science, Nanchang University, Nanchang, 330031, China
| | - Xiaofen Xie
- College of Life Science, Nanchang University, Nanchang, 330031, China
| | - Guoqin Qi
- College of Life Science, Nanchang University, Nanchang, 330031, China
| | - Panwei Weng
- College of Life Science, Nanchang University, Nanchang, 330031, China
| | - Zhizhen Hu
- College of Life Science, Nanchang University, Nanchang, 330031, China
| | - Kun Han
- College of Life Science, Nanchang University, Nanchang, 330031, China
| | - Huiwen Yuan
- College of Life Science, Nanchang University, Nanchang, 330031, China
| | - Yinping Li
- College of Life Science, Nanchang University, Nanchang, 330031, China
| | - Xiaoqin Ran
- College of Life Science, Nanchang University, Nanchang, 330031, China
| | - Gang Lin
- College of Life Science, Nanchang University, Nanchang, 330031, China.
| | - Chengyu Hu
- College of Life Science, Nanchang University, Nanchang, 330031, China.
| |
Collapse
|
168
|
Zhu Z, Li R, Wang L, Zheng Y, Hoque SAM, Lv Y, Zeng W. Glycogen Synthase Kinase-3 Regulates Sperm Motility and Acrosome Reaction via Affecting Energy Metabolism in Goats. Front Physiol 2019; 10:968. [PMID: 31417426 PMCID: PMC6682598 DOI: 10.3389/fphys.2019.00968] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 07/11/2019] [Indexed: 12/13/2022] Open
Abstract
Hyperactivation and acrosome reaction of sperm are pre-requisite steps for fertilization. However, the hyperactivation and acrosome reaction are critically controlled through the phosphorylation of specific proteins. Glycogen synthase kinase-3 (GSK3), a serine/threonine kinase with two different isoforms (α and β), is involved in biochemical signaling pathways. This study was aimed to investigate whether the GSK3α/β is present in goat sperm and its regulatory role in sperm motility and acrosome reaction. GSK3α/β was detected with immunofluorescence and Western blotting. Sperm motility, membrane integrity, acrosome reaction, mitochondrial membrane potential, phospho-Ser21-GSK3α and phospho-Ser9-GSK3β were analyzed. The ATP production and activities of lactate dehydrogenase (LDH), malate dehydrogenase (MDH), and succinate dehydrogenase (SDH) were measured. It was observed that the GSK3α/β was expressed in goat sperm, especially in the peri-acrosomal, mid-piece and principal piece of the tail. The abundance of GSK3α/β in sperm was increased during transit along the epididymis. Addition of either 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) or CHIR99021 significantly increased the sperm motility patterns and GSK3α/β phosphorylation. Interestingly, the adenosine triphosphate (ATP) production, activities of LDH, MDH and SDH were observed to be increased in the CHIR99021 treatment. The results suggested that GSK3α/β regulates sperm motility and acrosome reaction via phospho-ser21-GSK3α and phospho-ser9-GSK3β that involved in the regulation of sperm energy metabolism.
Collapse
Affiliation(s)
- Zhendong Zhu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Rongnan Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Liqiang Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Yi Zheng
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - S A Masudul Hoque
- Department of Animal Breeding and Genetics, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur, Bangladesh
| | - Yinghua Lv
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China.,College of Chemistry & Pharmacy, Northwest A&F University, Yangling, China
| | - Wenxian Zeng
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| |
Collapse
|
169
|
Hurcombe JA, Lay AC, Ni L, Barrington AF, Woodgett JR, Quaggin SE, Welsh GI, Coward RJ. Podocyte GSK3α is important for autophagy and its loss detrimental for glomerular function. FASEB Bioadv 2019; 1:498-510. [PMID: 31825015 PMCID: PMC6902909 DOI: 10.1096/fba.2019-00011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Podocytes are key cells in maintaining the integrity of the glomerular filtration barrier and preventing albuminuria. Glycogen synthase kinase 3 (GSK3) is a multi-functional serine/threonine kinase existing as two distinct but related isoforms (α and β). In the podocyte it has previously been reported that inhibition of the β isoform is beneficial in attenuating a variety of glomerular disease models but loss of both isoforms is catastrophic. However, it is not known what the role of GSK3α is in these cells. We now show that GSK3α is present and dynamically modulated in podocytes. When GSK3α is transgenically knocked down specifically in the podocytes of mice it causes mild but significant albuminuria by 6-weeks of life. Its loss also does not protect in models of diabetic or Adriamycin-induced nephropathy. In vitro deletion of podocyte GSK3α causes cell death and impaired autophagic flux suggesting it is important for this key cellular process. Collectively this work shows that GSK3α is important for podocyte health and that augmenting its function may be beneficial in treating glomerular disease.
Collapse
Affiliation(s)
| | - A C Lay
- Bristol Renal, University of Bristol
| | - L Ni
- Bristol Renal, University of Bristol
| | | | - J R Woodgett
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System & University of Toronto, Canada
| | - S E Quaggin
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois, US
| | - G I Welsh
- Bristol Renal, University of Bristol
| | | |
Collapse
|
170
|
Li H, Cao X, Chen X, Yi X, Xia J, Chen J, Yang L. Bufadienolides induce apoptosis and autophagy by inhibiting the AKT signaling pathway in melanoma A‑375 cells. Mol Med Rep 2019; 20:2347-2354. [PMID: 31322190 DOI: 10.3892/mmr.2019.10452] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 04/04/2019] [Indexed: 11/06/2022] Open
Abstract
The purpose of the present study was to investigate the effect of bufadienolides on the A‑375 melanoma cell line, and to delineate the underlying mechanism. A Cell Counting Kit‑8 assay was used to determine the viability of the cells, and flow cytometry was used to evaluate apoptosis. Western blot analysis was used to evaluate the expression levels of proteins involved in the AKT pathway that are associated with apoptosis and autophagy. The results demonstrated that bufadienolides reduced the viability of A‑375 cells in a dose‑ and a time‑dependent manner. Following treatment with bufadienolides, A‑375 cells exhibited clear properties that were characteristic of apoptosis and autophagy. The expression levels of the pro‑apoptotic proteins Bax and p53 were upregulated, whereas those of the anti‑apoptotic proteins, Bcl‑2 and caspase‑3 were downregulated. In addition, the level of a protein known to be associated with autophagy, microtubule‑associated proteins 1A/1B light chain 3‑II, was increased, whereas that of p62 protein was reduced. Finally, the AKT signaling pathway was blocked in the bufadienolide‑treated A‑375 cells. In conclusion, these results revealed that bufadienolides effectively induced apoptosis and autophagy in A‑375 cells via the AKT pathway, and therefore may be one of the candidate targets for the future development of targeted drugs to treat melanoma.
Collapse
Affiliation(s)
- Huannan Li
- Department of Anatomy, Histology and Embryology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, P.R. China
| | - Xiaoren Cao
- Department of Anatomy, Histology and Embryology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, P.R. China
| | - Xiongbing Chen
- Department of Anatomy, Histology and Embryology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, P.R. China
| | - Xiaohong Yi
- Department of Anatomy, Histology and Embryology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, P.R. China
| | - Jun Xia
- Department of Anatomy, Histology and Embryology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, P.R. China
| | - Jilan Chen
- Department of Anatomy, Histology and Embryology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, P.R. China
| | - Lan Yang
- Department of Anatomy, Histology and Embryology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, P.R. China
| |
Collapse
|
171
|
Magnesium lithospermate B protects the endothelium from inflammation-induced dysfunction through activation of Nrf2 pathway. Acta Pharmacol Sin 2019; 40:867-878. [PMID: 30617294 DOI: 10.1038/s41401-018-0189-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 10/18/2018] [Indexed: 12/22/2022]
Abstract
Magnesium lithospermate B (MLB) is an active component of Salvia miltiorrhiza Radix, a traditional Chinese herb used in treating cardiovascular diseases. In this study, we investigated the protective effects of MLB against inflammation-induced endothelial dysfunction in vitro and in vivo, and the underlying mechanisms. Endothelial dysfunction was induced in human dermal microvascular endothelial cells (HMEC-1) in vitro by lipopolysaccharide (LPS, 1 μg/mL). We showed that pretreatment with MLB (10-100 μM) dose-dependently inhibited LPS-induced upregulation of inflammatory cytokines ICAM1, VCAM1, and TNFα, which contributed to reduced leukocytes adhesion and attenuation of endothelial hyperpermeability in HMEC-1 cells. SD rats were injected with LPS (10 mg/kg, ip) to induce endothelial dysfunction in vivo. We showed that pretreatment with MLB (25-100 mg/kg, ip) dose-dependently restored LPS-impaired endothelial-dependent vasodilation in superior mesenteric artery (SMA), attenuated leukocyte adhesion in mesenteric venules and decreased vascular leakage in the lungs. We further elucidated the mechanisms underlying the protective effects of MLB, and revealed that MLB pretreatment inhibited NF-κB activation through inhibition of IκBα degradation and subsequent phosphorylation of NF-κB p65 in vitro and in vivo. In HMEC-1 cells, MLB pretreatment activated the nuclear factor erythroid-2-related factor 2 (Nrf2) pathway. Knockdown of Nrf2 with siRNA abolished the inhibitory effects of MLB on IκBα degradation and ICAM1 up-regulation, which were mimicked by PKC inhibition (Gö6983) or PI3K/Akt inhibition (LY294002). In summary, our results demonstrate that MLB inhibits NF-κB activation through PKC- and PI3K/Akt-mediated Nrf2 activation in HMEC-1 cells and protects against LPS-induced endothelial dysfunction in murine model of acute inflammation.
Collapse
|
172
|
Huang M, Zhang X, Li J, Li Y, Wang Q, Teng W. Comparison of osteogenic differentiation induced by siNoggin and pBMP-2 delivered by lipopolysaccharide-amine nanopolymersomes and underlying molecular mechanisms. Int J Nanomedicine 2019; 14:4229-4245. [PMID: 31239677 PMCID: PMC6559258 DOI: 10.2147/ijn.s203540] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 03/26/2019] [Indexed: 01/08/2023] Open
Abstract
Purpose: Gene therapies via Noggin small interfering (si)RNA (siNoggin) and bone morphogenetic protein (BMP)-2 plasmid DNA (pBMP-2) may be promising strategies for bone repair/regeneration, but their ideal delivery vectors, efficacy difference, and underlying mechanisms have not been explored, so these issues were probed here. Methods: This study used lipopolysaccharide-amine nanopolymersomes (LNPs), an efficient cytosolic delivery vector developed by the research team, to mediate siNoggin and pBMP-2 to transfect MC3T3-E1 cells, respectively. The cytotoxicity, cell uptake, and gene knockdown efficiency of siNoggin-loaded LNPs (LNPs/siNoggin) were studied, then the osteogenic-differentiation efficacy of MC3T3-E1 cells treated by LNPs/pBMP-2 and LNPs/siNoggin, respectively, were compared by measuring the expression of osteogenesis-related genes and proteins, alkaline phosphatase (ALP) activity, and mineralization of the extracellular matrix at all osteogenic stages. Finally, the possible signaling pathways of the two treatments were explored. Results: LNPs delivered siNoggin into cells efficiently to silence 50% of Noggin expression without obvious cytotoxicity. LNPs/siNoggin and LNPs/pBMP-2 enhanced the osteogenic differentiation of MC3T3 E1 cells, but LNPs/siNoggin was better than LNPs/pBMP-2. BMP/Mothers against decapentaplegic homolog (Smad) and glycogen synthase kinase (GSK)-3β/β-catenin signaling pathways appeared to be involved in osteogenic differentiation induced by LNPs/siNoggin, but GSK-3β/β-catenin was not stimulated upon LNPs/pBMP-2 treatment. Conclusion: LNPs are safe and efficient delivery vectors for DNA and RNA, which may find wide applications in gene therapy. siNoggin treatment may be a more efficient strategy to enhance osteogenic differentiation than pBMP-2 treatment. LNPs loaded with siNoggin and/or pBMP-2 may provide new opportunities for the repair and regeneration of bone.
Collapse
Affiliation(s)
- Mingdi Huang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Institute of Stomatological Research, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xinchun Zhang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Institute of Stomatological Research, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jing Li
- Laboratory of Biomaterials, Key Laboratory on Assisted Circulation, Ministry of Health, Cardiovascular Division, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yanshan Li
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Institute of Stomatological Research, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Qinmei Wang
- Laboratory of Biomaterials, Key Laboratory on Assisted Circulation, Ministry of Health, Cardiovascular Division, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Wei Teng
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Institute of Stomatological Research, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
173
|
Qu J, Yue L, Gao J, Yao H. Perspectives on Wnt Signal Pathway in the Pathogenesis and Therapeutics of Chronic Obstructive Pulmonary Disease. J Pharmacol Exp Ther 2019; 369:473-480. [PMID: 30952680 PMCID: PMC6538889 DOI: 10.1124/jpet.118.256222] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 04/04/2019] [Indexed: 12/16/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic lung disease with progressive airflow limitation and functional decline. The pathogenic mechanisms for this disease include oxidative stress, inflammatory responses, disturbed protease/antiprotease equilibrium, apoptosis/proliferation imbalance, senescence, autophagy, metabolic reprogramming, and mitochondrial dysfunction. The Wnt signaling pathway is an evolutionarily conserved signaling pathway that is abnormal in COPD, including chronic bronchitis and pulmonary emphysema. Furthermore, Wnt signaling has been shown to modulate aforementioned cellular processes involved in COPD. From this perspective, we provide an updated understanding of the crosstalk between Wnt signal and these cellular processes, and highlight the crucial role of the Wnt signal during the development of COPD. We also discuss the potential for targeting the Wnt signal in future translational and pharmacological therapeutics aimed at prevention and treatment of this disease.
Collapse
Affiliation(s)
- Jiao Qu
- The Second Affiliated Hospital, School of Pharmacy, Dalian Medical University, Dalian, Liaoning, China (J. Q., J. G.); The First Affiliated Hospital, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China (J.Q., J.G.); Department of Orthopedics, Warren Alpert Medical School, Brown University/Rhode Island Hospital, Providence, Rhode Island (L.Y.); and Department of Molecular Biology, Cell Biology and Biochemistry, Brown University Division of Biology and Medicine, Providence, Rhode Island (H.Y.)
| | - Li Yue
- The Second Affiliated Hospital, School of Pharmacy, Dalian Medical University, Dalian, Liaoning, China (J. Q., J. G.); The First Affiliated Hospital, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China (J.Q., J.G.); Department of Orthopedics, Warren Alpert Medical School, Brown University/Rhode Island Hospital, Providence, Rhode Island (L.Y.); and Department of Molecular Biology, Cell Biology and Biochemistry, Brown University Division of Biology and Medicine, Providence, Rhode Island (H.Y.)
| | - Jian Gao
- The Second Affiliated Hospital, School of Pharmacy, Dalian Medical University, Dalian, Liaoning, China (J. Q., J. G.); The First Affiliated Hospital, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China (J.Q., J.G.); Department of Orthopedics, Warren Alpert Medical School, Brown University/Rhode Island Hospital, Providence, Rhode Island (L.Y.); and Department of Molecular Biology, Cell Biology and Biochemistry, Brown University Division of Biology and Medicine, Providence, Rhode Island (H.Y.)
| | - Hongwei Yao
- The Second Affiliated Hospital, School of Pharmacy, Dalian Medical University, Dalian, Liaoning, China (J. Q., J. G.); The First Affiliated Hospital, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China (J.Q., J.G.); Department of Orthopedics, Warren Alpert Medical School, Brown University/Rhode Island Hospital, Providence, Rhode Island (L.Y.); and Department of Molecular Biology, Cell Biology and Biochemistry, Brown University Division of Biology and Medicine, Providence, Rhode Island (H.Y.)
| |
Collapse
|
174
|
Guo Q, Huang F, Goncalves C, Del Rincón SV, Miller WH. Translation of cancer immunotherapy from the bench to the bedside. Adv Cancer Res 2019; 143:1-62. [PMID: 31202357 DOI: 10.1016/bs.acr.2019.03.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
The tremendous success of immune checkpoint blockades has revolutionized cancer management. Our increased understanding of the cell types that compose the tumor microenvironment (TME), including those of the innate and adaptive immune system, has helped to shape additional immune modulatory strategies in cancer care. Pre-clinical and clinical investigations targeting novel checkpoint interactions and key pathways that regulate cancer immunity continue to increase rapidly. Various combinatorial drug regimens are being tested in attempt to achieve durable response and survival rates of patients with cancer. This review provides an overview of specific components of the TME, an introduction to novel immune checkpoints, followed by a survey of present day and future combination immune modulatory therapies. The idea that the immune system can recognize and destroy tumor cells was first described in the cancer immunosurveillance hypothesis of Burnet and Thomas. However, early experimental evidence failed to support the concept. It was not until the late 1990s when seminal papers clearly showed the existence of cancer immunosurveillance, leading to the cancer immunoediting hypothesis. In this century, progress in the understanding of negative regulators of the immune response led to the discovery that inhibition of these regulators in patients with cancer could lead to dramatic and durable remissions. Drs. Tasuku Honjo and James P. Allison were awarded the Nobel Prize in 2018 for their pioneering work in this field. We now see rapid advances in cancer immunology and emerging effective therapies revolutionizing cancer care across tumor types in the clinic, while pre-clinical research is moving from a focus on the malignant cells themselves to dissect the highly heterogenic and complex multi-cellular tumor microenvironment (TME).
Collapse
Affiliation(s)
- Qianyu Guo
- Division of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada; Jewish General Hospital, Segal Cancer Centre, Department of Oncology, Montreal, QC, Canada
| | - Fan Huang
- Division of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada; Jewish General Hospital, Segal Cancer Centre, Department of Oncology, Montreal, QC, Canada
| | - Christophe Goncalves
- Jewish General Hospital, Segal Cancer Centre, Department of Oncology, Montreal, QC, Canada
| | - Sonia V Del Rincón
- Division of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada; Jewish General Hospital, Segal Cancer Centre, Department of Oncology, Montreal, QC, Canada
| | - Wilson H Miller
- Division of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada; Jewish General Hospital, Segal Cancer Centre, Department of Oncology, Montreal, QC, Canada; Rossy Cancer Network, Montreal, QC, Canada.
| |
Collapse
|
175
|
Abstract
Glycogen synthase kinase-3 (GSK-3) is a cytoplasmic serine/threonine protein kinase which is known to regulate a variety of cellular processes through a number of signaling pathways important for cell proliferation, stem cell renewal, apoptosis and development. Although GSK-3 exists in a variety of tissues, this kinase plays very important roles in the heart to control its development through the formation of heart and cardiomyocyte proliferation. GSK-3 is also recognized as one of the main molecules that control cardiac hypertrophy and fibrosis. Therefore, GSK-3 could be an attractive target for the development of new drugs to cure cardiac diseases. The present review summarizes the roles of GSK-3 in the signaling pathways and the heart, and discusses the possibility of new drug development targeting this kinase.
Collapse
|
176
|
AKTivation mechanisms. Curr Opin Struct Biol 2019; 59:47-53. [PMID: 30901610 DOI: 10.1016/j.sbi.2019.02.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 02/06/2019] [Accepted: 02/08/2019] [Indexed: 12/21/2022]
Abstract
Akt1-3 (Akt) are a subset of the AGC protein Ser/Thr kinase family and play important roles in cell growth, metabolic regulation, cancer, and other diseases. We describe some of the roles of Akt in cell signaling and the biochemical and structural mechanisms of the regulation of Akt catalysis by the phospholipid PIP3 and by phosphorylation. Recent findings highlight a diverse set of strategies to control Akt catalytic activity to ensure its normal biological functions.
Collapse
|
177
|
Pavlov D, Bettendorff L, Gorlova A, Olkhovik A, Kalueff AV, Ponomarev ED, Inozemtsev A, Chekhonin V, Lesсh KP, Anthony DC, Strekalova T. Neuroinflammation and aberrant hippocampal plasticity in a mouse model of emotional stress evoked by exposure to ultrasound of alternating frequencies. Prog Neuropsychopharmacol Biol Psychiatry 2019; 90:104-116. [PMID: 30472146 DOI: 10.1016/j.pnpbp.2018.11.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 11/08/2018] [Accepted: 11/21/2018] [Indexed: 02/06/2023]
Abstract
Emotional stress is a form of stress evoked by processing negative mental experience rather than an organic or physical disturbance and is a frequent cause of neuropsychiatric pathologies, including depression. Susceptibility to emotional stress is commonly regarded as a human-specific trait that is challenging to model in other species. Recently, we showed that a 3-week-long exposure to ultrasound of unpredictable alternating frequencies within the ranges of 20-25 kHz and 25-45 kHz can induce depression-like characteristics in laboratory mice and rats. In an anti-depressant sensitive manner, exposure decreases sucrose preference, elevates behavioural despair, increases aggression, and alters serotonin-related gene expression. To further investigate this paradigm, we studied depression/distress-associated markers of neuroinflammation, neuroplasticity, oxidative stress and the activity of glycogen synthase kinase-3 (GSK-3) isoforms in the hippocampus of male mice. Stressed mice exhibited a decreased density of Ki67-positive and DCX-positive cells in the subgranular zone of hippocampus, and altered expression of brain-derived neurotrophic factor (BDNF), its receptor TrkB, and anti-apoptotic protein kinase B phosphorylated at serine 473 (AktpSer473). The mice also exhibited increased densities of Iba-1-positive cells, increased oxidative stress, increased levels of interleukin-1β (IL-1β), interleukin-6 (IL-6) in the hippocampus and plasma, and elevated activity of GSK-3 isoforms. Together, the results of our investigation have revealed that unpredictable alternating ultrasound evokes behavioural and molecular changes that are characteristic of the depressive syndrome and validates this new and simple method of modeling emotional stress in rodents.
Collapse
Affiliation(s)
- Dmitrii Pavlov
- Department of Neuroscience, Maastricht University, Universiteitssingel 40, NL 6229ER, Maastricht, Netherlands; Department of Biology, Lomonosov Moscow State University, Leninskie Gory1-12, Moscow 119991, Russia; Laboratory of Neurophysiology, GIGA-Neurosciences, University of Liège, Av. Hippocrate 15, Liège 4000, Belgium; Institute of General Pathology and Pathophysiology, Baltiiskaya str, 8, Moscow 125315, Russia
| | - Lucien Bettendorff
- Laboratory of Neurophysiology, GIGA-Neurosciences, University of Liège, Av. Hippocrate 15, Liège 4000, Belgium
| | - Anna Gorlova
- Department of Biology, Lomonosov Moscow State University, Leninskie Gory1-12, Moscow 119991, Russia; Laboratory of Neurophysiology, GIGA-Neurosciences, University of Liège, Av. Hippocrate 15, Liège 4000, Belgium; Sechenov First Moscow State Medical University, Institute of Molecular Medicine, Laboratory of Psychiatric Neurobiology and Department of Normal Physiology, Trubetskaya street 8-2, 119991, Moscow, Russia
| | - Andrey Olkhovik
- Department of Biology, Lomonosov Moscow State University, Leninskie Gory1-12, Moscow 119991, Russia
| | - Allan V Kalueff
- Institute of Translational Biomedicine, St.Petersburg State University, Universitetskaya nab. 7-9, St.-Petersburg 199034, Russia
| | - Eugene D Ponomarev
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Anatoly Inozemtsev
- Department of Biology, Lomonosov Moscow State University, Leninskie Gory1-12, Moscow 119991, Russia
| | - Vladimir Chekhonin
- Department of Basic and Applied Neurobiology, Serbsky Federal Medical Research Center for Psychiatry and Narcology, Kropotkinsky per 23, Moscow 119034, Russia
| | - Klaus-Peter Lesсh
- Department of Neuroscience, Maastricht University, Universiteitssingel 40, NL 6229ER, Maastricht, Netherlands; Sechenov First Moscow State Medical University, Institute of Molecular Medicine, Laboratory of Psychiatric Neurobiology and Department of Normal Physiology, Trubetskaya street 8-2, 119991, Moscow, Russia; Division of Molecular Psychiatry, Center of Mental Health University of Wuerzburg, Josef-Schneider-Straße 2, Wuerzburg 97080, Germany
| | - Daniel C Anthony
- Department of Pharmacology, Oxford University, Mansfield Road, Oxford OX1 3QT, UK.
| | - Tatyana Strekalova
- Department of Neuroscience, Maastricht University, Universiteitssingel 40, NL 6229ER, Maastricht, Netherlands; Institute of General Pathology and Pathophysiology, Baltiiskaya str, 8, Moscow 125315, Russia; Sechenov First Moscow State Medical University, Institute of Molecular Medicine, Laboratory of Psychiatric Neurobiology and Department of Normal Physiology, Trubetskaya street 8-2, 119991, Moscow, Russia.
| |
Collapse
|
178
|
Chen K, Lai K, Zhang X, Qin Z, Fu Q, Luo C, Jin X, Hu J, Liu S, Yao K. Bromfenac Inhibits TGF-β1-Induced Fibrotic Effects in Human Pterygium and Conjunctival Fibroblasts. Invest Ophthalmol Vis Sci 2019; 60:1156-1164. [PMID: 30908581 DOI: 10.1167/iovs.18-24743] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Nonsteroidal anti-inflammatory drugs (NSAIDs) have shown antifibrotic effects on several diseases. The aims of the present in vitro study were to investigate the antifibrotic effects of bromfenac (a kind of NSAID) on primary human pterygium fibroblasts (HPFs) and primary human conjunctival fibroblasts (HConFs), as well as to explore the possible mechanisms of these effects. Methods The cells used in this study were primary HPFs and HConFs, and profibrotic activation was induced by transforming growth factor-beta1 (TGF-β1). Western blot, quantitative real-time PCR, and immunofluorescence (IF) assays were used to detect the effects of TGF-β1 and bromfenac on the synthesis of fibronectin (FN), type III collagen (COL3), and alpha-smooth muscle actin (α-SMA) in HPFs and HConFs; the changes of signaling pathways were detected by Western blot; cell migration ability was detected by wound healing assay; cell proliferation ability was detected by CCK-8 assay; and pharmaceutical inhibitions of the downstream signaling pathways of TGF-β1 were used to assess their possible associations with the effects of bromfenac. Results Bromfenac suppressed the TGF-β1-induced protein expression of FN (0.59 ± 0.07 folds, P = 0.008), COL3 (0.48 ± 0.08 folds, P = 0.001), and α-SMA (0.61 ± 0.03 folds, P = 0.008) in HPFs. Bromfenac also attenuated TGF-β1-induced cell migration (0.30 ± 0.07 folds, P < 0.001), cell proliferation (0.64 ± 0.03 folds, P = 0.002) and the expression levels of p-AKT (0.66 ± 0.08 folds, P = 0.032), p-ERK1/2 (0.69 ± 0.11 folds, P = 0.003), and p-GSK-3β-S9 (0.65 ± 0.10 folds, P = 0.002) in HPFs. PI3K/AKT inhibitor (wortmannin) and MEK/ERK inhibitor (U0126) reduced the TGF-β1-induced synthesis of FN, COL3, and α-SMA in HPFs. All the results were similar in HConFs. Conclusions Bromfenac protects against TGF-β1-induced synthesis of FN, α-SMA, and COL3 in HPFs and HConFs at least in part by inactivating the AKT and ERK pathways.
Collapse
Affiliation(s)
- Kailin Chen
- Eye Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory of Ophthalmology of Zhejiang Province, Hangzhou, People's Republic of China
| | - Kairan Lai
- Eye Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory of Ophthalmology of Zhejiang Province, Hangzhou, People's Republic of China
| | - Xiaobo Zhang
- Eye Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory of Ophthalmology of Zhejiang Province, Hangzhou, People's Republic of China
| | - Zhenwei Qin
- Eye Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory of Ophthalmology of Zhejiang Province, Hangzhou, People's Republic of China
| | - Qiuli Fu
- Eye Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory of Ophthalmology of Zhejiang Province, Hangzhou, People's Republic of China
| | - Chenqi Luo
- Eye Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory of Ophthalmology of Zhejiang Province, Hangzhou, People's Republic of China
| | - Xiuming Jin
- Eye Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory of Ophthalmology of Zhejiang Province, Hangzhou, People's Republic of China
| | - Jianghua Hu
- Eye Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory of Ophthalmology of Zhejiang Province, Hangzhou, People's Republic of China
| | - Siyu Liu
- Eye Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory of Ophthalmology of Zhejiang Province, Hangzhou, People's Republic of China
| | - Ke Yao
- Eye Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory of Ophthalmology of Zhejiang Province, Hangzhou, People's Republic of China
| |
Collapse
|
179
|
Albrecht LV, Bui MH, De Robertis EM. Canonical Wnt is inhibited by targeting one-carbon metabolism through methotrexate or methionine deprivation. Proc Natl Acad Sci U S A 2019; 116:2987-2995. [PMID: 30679275 PMCID: PMC6386671 DOI: 10.1073/pnas.1820161116] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The nutrient-sensing metabolite S-adenosylmethionine (SAM) controls one-carbon metabolism by donating methyl groups to biochemical building blocks, DNA, RNA, and protein. Our recent work uncovered a requirement for cytoplasmic arginine methylation during Wnt signaling through the activity of protein arginine methyltransferase 1 (PRMT1), which transfers one-carbon groups from SAM to many protein substrates. Here, we report that treatments that decrease levels of the universal methyl donor SAM were potent inhibitors of Wnt signaling and of Wnt-induced digestion of extracellular proteins in endolysosomes. Thus, arginine methylation provides the canonical Wnt pathway with metabolic sensing properties through SAM. The rapid accumulation of Wnt-induced endolysosomes within 30 minutes was inhibited by the depletion of methionine, an essential amino acid that serves as the direct substrate for SAM production. We also found that methionine is required for GSK3 sequestration into multivesicular bodies through microautophagy, an essential step in Wnt signaling activity. Methionine starvation greatly reduced Wnt-induced endolysosomal degradation of extracellular serum proteins. Similar results were observed by addition of nicotinamide (vitamin B3), which serves as a methyl group sink. Methotrexate, a pillar in the treatment of cancer since 1948, decreases SAM levels. We show here that methotrexate blocked Wnt-induced endocytic lysosomal activity and reduced canonical Wnt signaling. Importantly, the addition of SAM during methionine depletion or methotrexate treatment was sufficient to rescue endolysosomal function and Wnt signaling. Inhibiting the Wnt signaling pathway by decreasing one-carbon metabolism provides a platform for designing interventions in Wnt-driven disease.
Collapse
Affiliation(s)
- Lauren V Albrecht
- Howard Hughes Medical Institute, University of California, Los Angeles, CA 90095-1662
- Department of Biological Chemistry, University of California, Los Angeles, CA 90095-1662
| | - Maggie H Bui
- Howard Hughes Medical Institute, University of California, Los Angeles, CA 90095-1662
- Department of Biological Chemistry, University of California, Los Angeles, CA 90095-1662
| | - Edward M De Robertis
- Howard Hughes Medical Institute, University of California, Los Angeles, CA 90095-1662;
- Department of Biological Chemistry, University of California, Los Angeles, CA 90095-1662
| |
Collapse
|
180
|
Arioka M, Takahashi-Yanaga F. Glycogen synthase kinase-3 inhibitor as a multi-targeting anti-rheumatoid drug. Biochem Pharmacol 2019; 165:207-213. [PMID: 30776323 DOI: 10.1016/j.bcp.2019.02.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 02/14/2019] [Indexed: 01/01/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory joint disease that causes swelling, bone erosion, and joint disorder. Patients with RA therefore suffer from pain and physiological disability, and have a decreased quality of life. During the progression of RA, many different types of cells and inflammatory factors influence each other with an important role. A better understanding of the pathology of RA should therefore lead to the development of effective anti-rheumatoid drugs, such as the anti-TNFα antibody. Glycogen synthase kinase-3 (GSK-3) is a cytoplasmic serine/threonine protein kinase that is involved in a large number of key cellular processes and is dysregulated in a wide variety of diseases, including inflammation and osteoporosis. The accumulated evidence has suggested that GSK-3 could be involved in multiple steps in the progression of RA. In the present review, the mechanisms of the pathogenesis of RA are summarized, and recent developments and potential new drugs targeting GSK-3 are discussed.
Collapse
Affiliation(s)
- Masaki Arioka
- Department of Clinical Pharmacology, Faculty of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Fumi Takahashi-Yanaga
- Department of Pharmacology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan.
| |
Collapse
|
181
|
Effects of chronic unpredictable mild stress induced prenatal stress on neurodevelopment of neonates: Role of GSK-3β. Sci Rep 2019; 9:1305. [PMID: 30718708 PMCID: PMC6361942 DOI: 10.1038/s41598-018-38085-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 12/13/2018] [Indexed: 11/12/2022] Open
Abstract
Prenatal stress (PNS) has gained attention with regard to its impact on hippocampal neurogenesis in neonates which serves as a risk factor for postnatal neurodevelopmental deficits. Evidences from animal models have suggested that depression responsive hypothalamic-pituitary-adrenal (HPA) axis and its hormonal response via cortisol, is responsible for critical neurodevelopmental deficits in the offspring which is transduced due to gestational stress. But knowledge in the area of assessing the effects of maternal chronic unpredictable mild stress (CUMS) on neurogenesis and expression of some key signaling molecules in the offsprings are limited. We have used Wistar rats to induce PNS in offsprings by maternal CUMS during pregnancy. Prefrontal cortex (PFC) and hippocampus were assessed for biomarkers of oxidative stress, neurogenesis, neurodevelopmental signaling molecules and DNA damage in the male Wister offsprings. Our investigations resulted in sufficient evidences which prove how maternal psychological stress has widespread effect on the fetal outcomes via major physiological alteration in the antioxidant levels, neurogenesis, signaling molecules and DNA damage. PNS leads to the upregulation of GSK-3β which in turn inhibited mRNA and protein expressions of sonic hedgehog (SHH), β-catenin, Notch and brain derived neurotrophic factor (BDNF). The study explored multifaceted signaling molecules especially, GSK-3β responsible for crosstalks between different neurodevelopmental molecules like SHH, Notch, BDNF and β-catenin affecting neurodevelopment of the offsprings due to PNS.
Collapse
|
182
|
Duan J, Cui J, Yang Z, Guo C, Cao J, Xi M, Weng Y, Yin Y, Wang Y, Wei G, Qiao B, Wen A. Neuroprotective effect of Apelin 13 on ischemic stroke by activating AMPK/GSK-3β/Nrf2 signaling. J Neuroinflammation 2019; 16:24. [PMID: 30709405 PMCID: PMC6357442 DOI: 10.1186/s12974-019-1406-7] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 01/11/2019] [Indexed: 01/01/2023] Open
Abstract
Background Previous studies had showed that Apelin 13 could protect against apoptosis induced by ischemic/reperfusion (I/R). However, the mechanisms whereby Apelin 13 protected brain I/R remained to be elucidated. The present study was designed to determine whether Apelin 13 provided protection through AMPK/GSK-3β/Nrf2 pathway. Methods In vivo, the I/R model was induced and Apelin 13 was given intracerebroventricularly 15 min before reperfusion. The neurobehavioral scores, infarction volumes, and some cytokines in the brain were measured. For in vitro study, PC12 cells were used. To clarify the mechanisms, proteases inhibitors or siRNA were used. Protein levels were investigated by western blotting. Results The results showed that Apelin 13 treatment significantly reduced infarct size, improved neurological outcomes, decreased brain edema, and inhibited cell apoptosis, oxidative stress, and neuroinflammation after I/R. Apelin 13 significantly increased the expression of Nrf2 and the phosphorylation levels of AMPK and GSK-3β. Furthermore, in cultured PC12 cells, the same protective effects were also observed. Silencing Nrf2 gene with its siRNA abolished the Apelin 13’s prevention of I/R-induced PC12 cell injury, oxidative stress, and inflammation. Inhibition of AMPK by its siRNA decreased the level of Apelin 13-induced Nrf2 expression and diminished the protective effects of Apelin 13. The interplay relationship between GSK-3β and Nrf2 was also verified with relative overexpression. Using selective inhibitors, we further identified the upstream of AMPK/GSK-3β/Nrf2 is AR/Gα/PLC/IP3/CaMKK. Conclusions In conclusion, the previous results showed that Apelin 13 protected against I/R-induced ROS-mediated inflammation and oxidative stress through activating the AMPK/GSK-3β pathway by AR/Gα/PLC/IP3/CaMKK signaling, and further upregulated the expression of Nrf2-regulated antioxidant enzymes.
Collapse
Affiliation(s)
- Jialin Duan
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, No. 127, Changle West Road, Xi'an, 710032, Shaanxi, China.,Department of Chinese Medicine, School of Life Science, Northwestern University, No. 229, Taibai Road, Xi'an, Shaanxi, China
| | - Jia Cui
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, No. 127, Changle West Road, Xi'an, 710032, Shaanxi, China
| | - Zhifu Yang
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, No. 127, Changle West Road, Xi'an, 710032, Shaanxi, China
| | - Chao Guo
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, No. 127, Changle West Road, Xi'an, 710032, Shaanxi, China
| | - Jinyi Cao
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, No. 127, Changle West Road, Xi'an, 710032, Shaanxi, China
| | - Miaomiao Xi
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, No. 127, Changle West Road, Xi'an, 710032, Shaanxi, China
| | - Yan Weng
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, No. 127, Changle West Road, Xi'an, 710032, Shaanxi, China
| | - Ying Yin
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, No. 127, Changle West Road, Xi'an, 710032, Shaanxi, China
| | - Yanhua Wang
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, No. 127, Changle West Road, Xi'an, 710032, Shaanxi, China
| | - Guo Wei
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, No. 127, Changle West Road, Xi'an, 710032, Shaanxi, China
| | - Boling Qiao
- Department of Chinese Medicine, School of Life Science, Northwestern University, No. 229, Taibai Road, Xi'an, Shaanxi, China.
| | - Aidong Wen
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, No. 127, Changle West Road, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
183
|
Hurcombe JA, Hartley P, Lay AC, Ni L, Bedford JJ, Leader JP, Singh S, Murphy A, Scudamore CL, Marquez E, Barrington AF, Pinto V, Marchetti M, Wong LF, Uney J, Saleem MA, Mathieson PW, Patel S, Walker RJ, Woodgett JR, Quaggin SE, Welsh GI, Coward RJM. Podocyte GSK3 is an evolutionarily conserved critical regulator of kidney function. Nat Commun 2019; 10:403. [PMID: 30679422 PMCID: PMC6345761 DOI: 10.1038/s41467-018-08235-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 12/21/2018] [Indexed: 01/18/2023] Open
Abstract
Albuminuria affects millions of people, and is an independent risk factor for kidney failure, cardiovascular morbidity and death. The key cell that prevents albuminuria is the terminally differentiated glomerular podocyte. Here we report the evolutionary importance of the enzyme Glycogen Synthase Kinase 3 (GSK3) for maintaining podocyte function in mice and the equivalent nephrocyte cell in Drosophila. Developmental deletion of both GSK3 isoforms (α and β) in murine podocytes causes late neonatal death associated with massive albuminuria and renal failure. Similarly, silencing GSK3 in nephrocytes is developmentally lethal for this cell. Mature genetic or pharmacological podocyte/nephrocyte GSK3 inhibition is also detrimental; producing albuminuric kidney disease in mice and nephrocyte depletion in Drosophila. Mechanistically, GSK3 loss causes differentiated podocytes to re-enter the cell cycle and undergo mitotic catastrophe, modulated via the Hippo pathway but independent of Wnt-β-catenin. This work clearly identifies GSK3 as a critical regulator of podocyte and hence kidney function.
Collapse
Affiliation(s)
- J A Hurcombe
- Bristol Renal, Dorothy Hodgkin Building, University of Bristol, Bristol, BS1 3NY, UK
| | - P Hartley
- Bournemouth University, Bournemouth, BH12 5BB, UK
| | - A C Lay
- Bristol Renal, Dorothy Hodgkin Building, University of Bristol, Bristol, BS1 3NY, UK
| | - L Ni
- Bristol Renal, Dorothy Hodgkin Building, University of Bristol, Bristol, BS1 3NY, UK
| | - J J Bedford
- Dunedin School of Medicine, University of Otago, Dunedin, 9016, New Zealand
| | - J P Leader
- Dunedin School of Medicine, University of Otago, Dunedin, 9016, New Zealand
| | - S Singh
- Bristol Renal, Dorothy Hodgkin Building, University of Bristol, Bristol, BS1 3NY, UK
| | - A Murphy
- Department of Pathology, Southern General Hospital, Glasgow, G51 4TF, UK
| | - C L Scudamore
- Mary Lyon Centre, MRC Harwell, Didcot, Oxford, OX11 0RD, UK
| | - E Marquez
- Bristol Renal, Dorothy Hodgkin Building, University of Bristol, Bristol, BS1 3NY, UK
| | - A F Barrington
- Bristol Renal, Dorothy Hodgkin Building, University of Bristol, Bristol, BS1 3NY, UK
| | - V Pinto
- Bristol Renal, Dorothy Hodgkin Building, University of Bristol, Bristol, BS1 3NY, UK
| | - M Marchetti
- Bristol Renal, Dorothy Hodgkin Building, University of Bristol, Bristol, BS1 3NY, UK
| | - L-F Wong
- Translational Health Sciences, University of Bristol, Bristol, BS2 8DZ, UK
| | - J Uney
- Translational Health Sciences, University of Bristol, Bristol, BS2 8DZ, UK
| | - M A Saleem
- Bristol Renal, Dorothy Hodgkin Building, University of Bristol, Bristol, BS1 3NY, UK
| | - P W Mathieson
- Bristol Renal, Dorothy Hodgkin Building, University of Bristol, Bristol, BS1 3NY, UK
- The University of Hong Kong, Pokfulam, Hong Kong
| | - S Patel
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System & University of Toronto, Toronto, M5G 1X5, Canada
- Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - R J Walker
- Dunedin School of Medicine, University of Otago, Dunedin, 9016, New Zealand
| | - J R Woodgett
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System & University of Toronto, Toronto, M5G 1X5, Canada
| | - S E Quaggin
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, 60611, IL, USA
| | - G I Welsh
- Bristol Renal, Dorothy Hodgkin Building, University of Bristol, Bristol, BS1 3NY, UK
| | - R J M Coward
- Bristol Renal, Dorothy Hodgkin Building, University of Bristol, Bristol, BS1 3NY, UK.
| |
Collapse
|
184
|
Klein MO, Battagello DS, Cardoso AR, Hauser DN, Bittencourt JC, Correa RG. Dopamine: Functions, Signaling, and Association with Neurological Diseases. Cell Mol Neurobiol 2019; 39:31-59. [PMID: 30446950 PMCID: PMC11469830 DOI: 10.1007/s10571-018-0632-3] [Citation(s) in RCA: 556] [Impact Index Per Article: 92.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 11/02/2018] [Indexed: 02/07/2023]
Abstract
The dopaminergic system plays important roles in neuromodulation, such as motor control, motivation, reward, cognitive function, maternal, and reproductive behaviors. Dopamine is a neurotransmitter, synthesized in both central nervous system and the periphery, that exerts its actions upon binding to G protein-coupled receptors. Dopamine receptors are widely expressed in the body and function in both the peripheral and the central nervous systems. Dopaminergic signaling pathways are crucial to the maintenance of physiological processes and an unbalanced activity may lead to dysfunctions that are related to neurodegenerative diseases. Unveiling the neurobiology and the molecular mechanisms that underlie these illnesses may contribute to the development of new therapies that could promote a better quality of life for patients worldwide. In this review, we summarize the aspects of dopamine as a catecholaminergic neurotransmitter and discuss dopamine signaling pathways elicited through dopamine receptor activation in normal brain function. Furthermore, we describe the potential involvement of these signaling pathways in evoking the onset and progression of some diseases in the nervous system, such as Parkinson's, Schizophrenia, Huntington's, Attention Deficit and Hyperactivity Disorder, and Addiction. A brief description of new dopaminergic drugs recently approved and under development treatments for these ailments is also provided.
Collapse
Affiliation(s)
- Marianne O Klein
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, 05508-000, Brazil
| | - Daniella S Battagello
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, 05508-000, Brazil
| | - Ariel R Cardoso
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, 05508-000, Brazil
| | - David N Hauser
- Center for Translational Neuroscience, Sanford Burnham Prebys (SBP) Medical Discovery Institute, 10901 North Torrey Pines Rd., La Jolla, CA, 92037, USA
| | - Jackson C Bittencourt
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, 05508-000, Brazil.
- Center for Neuroscience and Behavior, Institute of Psychology, USP, São Paulo, Brazil.
| | - Ricardo G Correa
- Center for Translational Neuroscience, Sanford Burnham Prebys (SBP) Medical Discovery Institute, 10901 North Torrey Pines Rd., La Jolla, CA, 92037, USA.
| |
Collapse
|
185
|
Abstract
SIGNIFICANCE Nuclear factor E2-related factor 2 (Nrf2) is a transcription factor that coordinates the basal and stress-inducible activation of a vast array of cytoprotective genes. Understanding the regulation of Nrf2 activity and downstream pathways has major implications for human health. Recent Advances: Nrf2 regulates the transcription of components of the glutathione and thioredoxin antioxidant systems, as well as enzymes involved in phase I and phase II detoxification of exogenous and endogenous products, NADPH regeneration, and heme metabolism. It therefore represents a crucial regulator of the cellular defense mechanisms against xenobiotic and oxidative stress. In addition to antioxidant responses, Nrf2 is involved in other cellular processes, such as autophagy, intermediary metabolism, stem cell quiescence, and unfolded protein response. Given the wide range of processes that Nrf2 controls, its activity is tightly regulated at multiple levels. Here, we review the different modes of regulation of Nrf2 activity and the current knowledge of Nrf2-mediated transcriptional control. CRITICAL ISSUES It is now clear that Nrf2 lies at the center of a complex regulatory network. A full comprehension of the Nrf2 program will require an integrated consideration of all the different factors determining Nrf2 activity. FUTURE DIRECTIONS Additional computational and experimental studies are needed to obtain a more dynamic global view of Nrf2-mediated gene regulation. In particular, studies comparing how the Nrf2-dependent network changes from a physiological to a pathological condition can provide insight into mechanisms of disease and instruct new treatment strategies.
Collapse
Affiliation(s)
- Claudia Tonelli
- 1 Cold Spring Harbor Laboratory , Cold Spring Harbor, New York
| | | | - David A Tuveson
- 1 Cold Spring Harbor Laboratory , Cold Spring Harbor, New York.,2 Lustgarten Foundation Pancreatic Cancer Research Laboratory , Cold Spring Harbor, New York
| |
Collapse
|
186
|
Chang H, Chang CY, Lee HJ, Chou CY, Chou TC. Magnolol ameliorates pneumonectomy and monocrotaline-induced pulmonary arterial hypertension in rats through inhibition of angiotensin II and endothelin-1 expression. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 51:205-213. [PMID: 30466619 DOI: 10.1016/j.phymed.2018.10.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 10/01/2018] [Accepted: 10/03/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Magnolol, a major bioactive component extracted from Magnolia officinalis, exerts several beneficial effects, such as anti-inflammatory and anti-hypertensive activities. PURPOSE In this study, we investigated whether magnolol has a protective effect on pneumonectomy and monocrotaline-induced pulmonary arterial hypertension (PAH) in rats. DESIGN/METHODS The alterations of right ventricular (RV) hypertrophy, pulmonary vascular remodeling, histopathological parameters, and related gene expression and signaling pathways in lungs by magnolol treatment were studied in the PAH rats. RESULTS Administration of magnolol greatly ameliorated the characteristic features of PAH, including increased pulmonary arterial pressure, RV hypertrophy, and pulmonary vascular remodeling. Moreover, magnolol inhibited angiotensin-converting enzyme (ACE)/angiotensin II (Ang II)/Ang II type 1 receptor (AT-1R) cascade, whereas upregulates ACE2 in the lungs of PAH rats. The overexpression of endothelin-1 (ET-1) and ETA receptor occurred in the PAH rats was significantly attenuated by magnolol through inhibition of Akt/ERK1/2/GSK3β/β-catenin pathway. Compared with that of untreated PAH rats, higher expression of endothelial nitric oxide synthase, and lower expression of inducible nitric oxide synthase and O2- production in lungs were observed in magnolol-treated PAH rats. CONCLUSION We demonstrated that treatment with magnolol reduces the development of PAH induced by pneumonectomy and monocrotaline in rats, and suppressing Ang II and ET-1-mediated processes may contribute to its protective effects. These findings suggest that magnolol may be a potential agent for PAH therapy.
Collapse
Affiliation(s)
- Hung Chang
- Division of Thoracic Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Cheng-Yi Chang
- Graduate Institute of Physiology, National Defense Medical Center, Taipei, Taiwan; Department of Respiratory Therapy, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Hwei-Jen Lee
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | - Ching-Yu Chou
- Graduate Institute of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | - Tz-Chong Chou
- Department of Medical Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan; Department of Biotechnology, Asia University, Taichung, Taiwan; China Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
187
|
Urbanska M, Kazmierska-Grebowska P, Kowalczyk T, Caban B, Nader K, Pijet B, Kalita K, Gozdz A, Devijver H, Lechat B, Jaworski T, Grajkowska W, Sadowski K, Jozwiak S, Kotulska K, Konopacki J, Van Leuven F, van Vliet EA, Aronica E, Jaworski J. GSK3β activity alleviates epileptogenesis and limits GluA1 phosphorylation. EBioMedicine 2018; 39:377-387. [PMID: 30502054 PMCID: PMC6355642 DOI: 10.1016/j.ebiom.2018.11.040] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 11/16/2018] [Accepted: 11/16/2018] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND Glycogen synthase kinase-3β (GSK3β) is a key regulator of cellular homeostasis. In neurons, GSK3β contributes to the control of neuronal transmission and plasticity, but its role in epilepsy remains to be defined. METHODS Biochemical and electrophysiological methods were used to assess the role of GSK3β in regulating neuronal transmission and epileptogenesis. GSK3β activity was increased genetically in GSK3β[S9A] mice. Its effects on neuronal transmission and epileptogenesis induced by kainic acid were assessed by field potential recordings in mice brain slices and video electroencephalography in vivo. The ion channel expression was measured in brain samples from mice and followed by analysis in samples from patients with temporal lobe epilepsy or focal cortical dysplasia in correlation to GSK3β phosphorylation. FINDINGS Higher GSK3β activity decreased the progression of kainic acid induced epileptogenesis. At the biochemical level, higher GSK3β activity increased the expression of hyperpolarization-activated cyclic nucleotide-gated (HCN) channel 4 under basal conditions and in the epileptic mouse brain and decreased phosphorylation of the glutamate α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunit GluA1 at Serine 831 under basal conditions. Moreover, we found a significant correlation between higher inhibitory GSK3β phosphorylation at Serine 9 and higher activating GluA1 phosphorylation at Serine 845 in brain samples from epileptic patients. INTERPRETATION Our data imply GSK3β activity in the protection of neuronal networks from hyper-activation in response to epileptogenic stimuli and indicate that the anti-epileptogenic function of GSK3β involves modulation of HCN4 level and the synaptic AMPA receptors pool.
Collapse
Affiliation(s)
- Malgorzata Urbanska
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology, Warsaw 02-109, Poland; Department of Neurology and Epileptology, Children's Memorial Health Institute, Warsaw 04-730, Poland.
| | - Paulina Kazmierska-Grebowska
- Department of Neurobiology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz 90-236, Poland
| | - Tomasz Kowalczyk
- Department of Neurobiology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz 90-236, Poland
| | - Bartosz Caban
- Department of Neurobiology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz 90-236, Poland
| | - Karolina Nader
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Science, Warsaw 02-093, Poland
| | - Barbara Pijet
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Science, Warsaw 02-093, Poland
| | - Katarzyna Kalita
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Science, Warsaw 02-093, Poland
| | - Agata Gozdz
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology, Warsaw 02-109, Poland
| | - Herman Devijver
- Experimental Genetics Group - LEGTEGG, Department of Human Genetics, KULeuven, Leuven 3000, Belgium
| | - Benoit Lechat
- Experimental Genetics Group - LEGTEGG, Department of Human Genetics, KULeuven, Leuven 3000, Belgium
| | - Tomasz Jaworski
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Science, Warsaw 02-093, Poland
| | - Wieslawa Grajkowska
- Department of Pathology, Children's Memorial Health Institute, Warsaw 04-730, Poland
| | - Krzysztof Sadowski
- Department of Neurology and Epileptology, Children's Memorial Health Institute, Warsaw 04-730, Poland
| | - Sergiusz Jozwiak
- Department of Neurology and Epileptology, Children's Memorial Health Institute, Warsaw 04-730, Poland; Department of Child Neurology, Medical University of Warsaw, Warsaw 02-091, Poland
| | - Katarzyna Kotulska
- Department of Neurology and Epileptology, Children's Memorial Health Institute, Warsaw 04-730, Poland
| | - Jan Konopacki
- Department of Neurobiology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz 90-236, Poland
| | - Fred Van Leuven
- Experimental Genetics Group - LEGTEGG, Department of Human Genetics, KULeuven, Leuven 3000, Belgium
| | - Erwin A van Vliet
- Amsterdam UMC, University of Amsterdam, Department of (Neuro) Pathology, Amsterdam Neuroscience, Amsterdam, the Netherlands; Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, 1098 XH, the Netherlands
| | - Eleonora Aronica
- Amsterdam UMC, University of Amsterdam, Department of (Neuro) Pathology, Amsterdam Neuroscience, Amsterdam, the Netherlands; Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, the Netherlands
| | - Jacek Jaworski
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology, Warsaw 02-109, Poland.
| |
Collapse
|
188
|
Pacureanu L, Avram S, Bora A, Kurunczi L, Crisan L. Portraying the selectivity of GSK-3 inhibitors towards CDK-2 by 3D similarity and molecular docking. Struct Chem 2018. [DOI: 10.1007/s11224-018-1224-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
189
|
Varghese E, Samuel SM, Abotaleb M, Cheema S, Mamtani R, Büsselberg D. The "Yin and Yang" of Natural Compounds in Anticancer Therapy of Triple-Negative Breast Cancers. Cancers (Basel) 2018; 10:E346. [PMID: 30248941 PMCID: PMC6209965 DOI: 10.3390/cancers10100346] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 09/18/2018] [Accepted: 09/19/2018] [Indexed: 02/07/2023] Open
Abstract
Among the different types of breast cancers, triple-negative breast cancers (TNBCs) are highly aggressive, do not respond to conventional hormonal/human epidermal growth factor receptor 2 (HER2)-targeted interventions due to the lack of the respective receptor targets, have chances of early recurrence, metastasize, tend to be more invasive in nature, and develop drug resistance. The global burden of TNBCs is increasing regardless of the number of cytotoxic drugs being introduced into the market each year as they have only moderate efficacy and/or unforeseen side effects. Therefore, the demand for more efficient therapeutic interventions, with reduced side effects, for the treatment of TNBCs is rising. While some plant metabolites/derivatives actually induce the risk of cancers, many plant-derived active principles have gained attention as efficient anticancer agents against TNBCs, with fewer adverse side effects. Here we discuss the possible oncogenic molecular pathways in TNBCs and how the purified plant-derived natural compounds specifically target and modulate the genes and/or proteins involved in these aberrant pathways to exhibit their anticancer potential. We have linked the anticancer potential of plant-derived natural compounds (luteolin, chalcones, piperine, deguelin, quercetin, rutin, fisetin, curcumin, resveratrol, and others) to their ability to target multiple dysregulated signaling pathways (such as the Wnt/β-catenin, Notch, NF-κB, PI3K/Akt/mammalian target of rapamycin (mTOR), mitogen-activated protein kinase (MAPK) and Hedgehog) leading to suppression of cell growth, proliferation, migration, inflammation, angiogenesis, epithelial-mesenchymal transition (EMT) and metastasis, and activation of apoptosis in TNBCs. Plant-derived compounds in combination with classical chemotherapeutic agents were more efficient in the treatment of TNBCs, possibly with lesser side effects.
Collapse
Affiliation(s)
- Elizabeth Varghese
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar.
| | - Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar.
| | - Mariam Abotaleb
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar.
| | - Sohaila Cheema
- Institute for Population Health, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar.
| | - Ravinder Mamtani
- Institute for Population Health, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar.
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar.
| |
Collapse
|
190
|
Vaseghi S, Babapour V, Nasehi M, Zarrindast MR. The role of CA1 CB1 receptors on lithium-induced spatial memory impairment in rats. EXCLI JOURNAL 2018; 17:916-934. [PMID: 30564071 PMCID: PMC6295625 DOI: 10.17179/excli2018-1511] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 08/27/2018] [Indexed: 02/06/2023]
Abstract
Lithium, a glycogen synthase kinase-3β (GSK-3β) inhibitor, prevents cannabinoid withdrawal syndrome, but there is limited data exploring the interaction between lithium and cannabinoid system on memory processes. The present study aimed to test the interaction between dorsal hippocampal (CA1 region) cannabinoid system and lithium on spatial memory in rats. Spatial memory was assessed in Morris Water Maze (MWM) apparatus by a single training session of eight trials. The results showed that pre-training intra-CA1 microinjection of ACPA, the cannabinoid type 1 receptor (CB1r) agonist, at doses of 0.001, 0.01 or 1 µg/rat, or AM251, the cannabinoid type 1 receptor (CB1r) antagonist, at doses of 1, 10 or 100 ng/rat, increased escape latency and traveled distance to the platform, suggesting a spatial learning impairment, whereas intraperitoneal administration of lithium (0.5, 1 or 5 mg/kg) had no effect on spatial learning. Also, rats that received lithium plus a lower dose of ACPA (0.001 µg/rat) or AM251 (1 ng/rat) had successful performance in the MWM. In the probe test, the results showed that pre-training administration of lithium (5 mg/kg) and ACPA (0.01 or 1 µg/rat) but not AM251 (at all doses used) impaired spatial memory retrieval. Also, lower dose of ACPA (0.001 µg/rat) or AM251 (1 ng/rat) potentiated the effect of ineffective doses of lithium (0.5 and 1 mg/kg) on spatial memory retrieval, while restored the effect of effective dose of lithium (5 mg/kg). In conclusion, cannabinoids may have a dual effect on lithium-induced spatial memory impairment in rats.
Collapse
Affiliation(s)
- Salar Vaseghi
- Department of Physiology, Faculty of Veterinary Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Vahab Babapour
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mohammad Nasehi
- Cognitive and Neuroscience Research Center (CNRC), Amir-Almomenin Hospital, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad-Reza Zarrindast
- Department of Pharmacology School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Institute for Cognitive Science Studies (ICSS), Tehran, Iran.,Department of Neuroendocrinology, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
191
|
Tan Z, Kang T, Zhang X, Tong Y, Chen S. Nerve growth factor prevents arsenic-induced toxicity in PC12 cells through the AKT/GSK-3β/NFAT pathway. J Cell Physiol 2018; 234:4726-4738. [PMID: 30256405 DOI: 10.1002/jcp.27255] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 07/25/2018] [Indexed: 12/13/2022]
Abstract
The potential risk of arsenic-related neurodegeneration has been a growing concern. Arsenic exposure has been reported to disrupt neurite growth and neuron body integrity in vitro; however, its underlying mechanism remains unclear. Previously, we showed that arsenic sulfide (AS) exerted cytotoxicity in gastric and colon cancer cells through regulating nuclear factor of the activated T cells (NFAT) pathway. The NFAT pathway regulates axon path finding and neural development. Using neural crest cell line PC12 cells as a model, here we show that AS caused mitochondrial membrane potential collapse, reactive oxygen species production, and cytochrome c release, leading to mitochondria-mediated apoptosis via the AKT/GSK-3β/NFAT pathway. Increased glycogen synthase kinase-3 beta (GSK-3β) activation leads to the inactivation of NFAT and its antiapoptotic effects. Through inhibiting GSK-3β activity, both nerve growth factor (NGF) and Tideglusib, a GSK-3β inhibitor partially rescued the PC12 cells from the AS-induced cytotoxicity and restored the expression of NFATc3. In addition, overexpression of NFATc3 stimulated neurite outgrowth and potentiated the effect of NGF on promoting the neurite outgrowth. Collectively, our results show that NFATc3 serves as the downstream target of NGF and plays a key role in preventing AS-induced neurotoxicity through regulating the AKT/GSK-3β/NFAT pathway in PC12 cells.
Collapse
Affiliation(s)
- Zhen Tan
- Department of Oncology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ting Kang
- Department of Oncology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiuli Zhang
- Department of Oncology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yingying Tong
- Department of Oncology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Siyu Chen
- Department of Oncology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
192
|
Li Q, Liao C, Xu W, Li G, Hong K, Cheng X, Li J. Xeroderma Pigmentosum Group D (XPD) Inhibits the Proliferation Cycle of Vascular Smooth Muscle Cell (VSMC) by Activating Glycogen Synthase Kinase 3β (GSK3β). Med Sci Monit 2018; 24:5951-5959. [PMID: 30146633 PMCID: PMC6122044 DOI: 10.12659/msm.909614] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND VSMC proliferation plays a key role in atherosclerosis, but the role of XPD in VSMC proliferation remains unknown. We investigated the expression of XPD, which is involved in cell cycle regulation, and its role in VSMC proliferation response to atherogenic stimuli. MATERIAL AND METHODS Human umbilical vein VSMCs were transfected with recombinant plasmid pEGFP-N2/XPD and pEGFP-N2 and incubated with PDGF-BB in vitro. Cell viability was determined by MTT assay. The expressions of XPD, GSK3β, p-GSK3β, CDK4, and cyclin D1 protein were detected by Western blot analysis. Cell cycle was examined by flow cytometry. RESULTS PDGF inhibited the expression of XPD in VSMCs and promoted VSMC proliferation. Overexpression of XPD significantly augmented cell cycle arrest, and attenuated protein expression levels of CDK4 and cyclin D1 in VSMCs. XPD overexpression suppressed the effects of PDGF-BB in promoting G1/S transition and accelerating protein expression levels of CDK4 and cyclin D1. XPD diminished the phosphorylation of GSK3β, and SB216763 inhibited the reduction effect of XPD on CDK4 and cyclin D1. CONCLUSIONS XPD induces VSMC cell cycle arrest, and the activation of GSK3β plays a crucial role in inhibitory effect of XPD on VSMC proliferation.
Collapse
Affiliation(s)
- Qing Li
- Department of Cardiology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland).,Jiangxi Provincial Key Laboratory of Molecular Medicine, Nanchang, Jiangxi, China (mainland)
| | - Chunyao Liao
- Department of Cardiology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Wang Xu
- Department of Cardiology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Genlin Li
- Department of Cardiology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Kui Hong
- Department of Cardiology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Xiaoshu Cheng
- Department of Cardiology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Juxiang Li
- Department of Cardiology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| |
Collapse
|
193
|
Liu X, Klein PS. Glycogen synthase kinase-3 and alternative splicing. WILEY INTERDISCIPLINARY REVIEWS-RNA 2018; 9:e1501. [PMID: 30118183 DOI: 10.1002/wrna.1501] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 07/03/2018] [Accepted: 07/09/2018] [Indexed: 12/16/2022]
Abstract
Glycogen synthase kinase-3 (GSK-3) is a highly conserved negative regulator of receptor tyrosine kinase, cytokine, and Wnt signaling pathways. Stimulation of these pathways inhibits GSK-3 to modulate diverse downstream effectors that include transcription factors, nutrient sensors, glycogen synthesis, mitochondrial function, circadian rhythm, and cell fate. GSK-3 also regulates alternative splicing in response to T-cell receptor activation, and recent phosphoproteomic studies have revealed that multiple splicing factors and regulators of RNA biosynthesis are phosphorylated in a GSK-3-dependent manner. Furthermore, inhibition of GSK-3 alters the splicing of hundreds of mRNAs, indicating a broad role for GSK-3 in the regulation of RNA processing. GSK-3-regulated phosphoproteins include SF3B1, SRSF2, PSF, RBM8A, nucleophosmin 1 (NPM1), and PHF6, many of which are mutated in leukemia and myelodysplasia. As GSK-3 is inhibited by pathways that are pathologically activated in leukemia and loss of Gsk3 in hematopoietic cells causes a severe myelodysplastic neoplasm in mice, these findings strongly implicate GSK-3 as a critical regulator of mRNA processing in normal and malignant hematopoiesis. This article is categorized under: RNA Processing > Splicing Mechanisms RNA Processing > Splicing Regulation/Alternative Splicing RNA in Disease and Development > RNA in Disease RNA Interactions with Proteins and Other Molecules > Protein-RNA Interactions: Functional Implications.
Collapse
Affiliation(s)
- Xiaolei Liu
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Peter S Klein
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
194
|
Challenges and perspectives in the treatment of diabetes associated breast cancer. Cancer Treat Rev 2018; 70:98-111. [PMID: 30130687 DOI: 10.1016/j.ctrv.2018.08.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 08/02/2018] [Accepted: 08/09/2018] [Indexed: 12/12/2022]
Abstract
Type 2 diabetes mellitus is one of the most common chronic disease worldwide and affects all cross-sections of the society including children, women, youth and adults. Scientific evidence has linked diabetes to higher incidence, accelerated progression and increased aggressiveness of different cancers. Among the different forms of cancer, research has reinforced a link between diabetes and the risk of breast cancer. Some studies have specifically linked diabetes to the highly aggressive, triple negative breast cancers (TNBCs) which do not respond to conventional hormonal/HER2 targeted interventions, have chances of early recurrence, metastasize, tend to be more invasive in nature and develop drug resistance. Commonly used anti-diabetic drugs, such as metformin, have recently gained importance in the treatment of breast cancer due to their proposed anti-cancer properties. Here we discuss the link between diabetes and breast cancer, the metabolic disturbances in diabetes that support the development of breast cancer, the challenges involved and future perspective and directions. We link the three main metabolic disturbances (dyslipidemia, hyperinsulinemia and hyperglycemia) that occur in diabetes to potential aberrant molecular pathways that may lead to the development of an oncogenic phenotype of the breast tissue, thereby leading to acceleration of cell growth, proliferation, migration, inflammation, angiogenesis, EMT and metastasis and inhibition of apoptosis in breast cancer cells. Furthermore, managing diabetes and treating cancer using a combination of anti-diabetic and classical anti-cancer drugs should prove to be more efficient in the treatment diabetes associated cancers.
Collapse
|
195
|
Bruchhage KL, Koennecke M, Drenckhan M, Plötze-Martin K, Pries R, Wollenberg B. 1,8-cineol inhibits the Wnt/β-catenin signaling pathway through GSK-3 dephosphorylation in nasal polyps of chronic rhinosinusitis patients. Eur J Pharmacol 2018; 835:140-146. [PMID: 30081034 DOI: 10.1016/j.ejphar.2018.07.060] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 07/25/2018] [Accepted: 07/31/2018] [Indexed: 12/17/2022]
Abstract
Chronic rhinosinusitis with nasal polyps (CRSwNP) represents a benign neoplasm of the nasal mucosa, which leads to a decreased breathing capacity and reduced olfaction. The pathogenesis and the molecular mechanisms driving nasal polyps are not very well known. GSK-3 is involved in the regulation of various biosynthetic pathways and various kinases are able to regulate the GSK-3. Therefore, we investigated the effect of the monoterpene oxide 1,8-cineol on the regulation of the Wnt/β-catenin signaling pathway with its central regulator protein GSK-3 in vitro. We determined GSK-3 expression and phosphorylation as well as the expression of negative regulators (Akt and SGK) and downstream activation of β-catenin in nasal polyps of patients with CRSwNP by immunohistochemistry and Western blot experiments. In this study we demonstrated for the first time, that 1,8-cineol acts as a potential inhibitor of the Wnt/β-catenin signaling pathway, by affecting the inhibitory phosphorylation of GSK-3, which is the key regulator of the β-catenin activity. Our data provide novel insights in the regulatory networks responsible for the progression of CRSwNP and furthermore represent a new mechanism of 1,8-cineol activity, which may lead to novel treatment approaches to this natural drug.
Collapse
Affiliation(s)
- Karl-Ludwig Bruchhage
- Department of Otorhinolaryngology, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Michael Koennecke
- Department of Otorhinolaryngology, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Maren Drenckhan
- Department of Otorhinolaryngology, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Kirstin Plötze-Martin
- Department of Otorhinolaryngology, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Ralph Pries
- Department of Otorhinolaryngology, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Barbara Wollenberg
- Department of Otorhinolaryngology, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany.
| |
Collapse
|
196
|
Zhou H, Wang H, Ni M, Yue S, Xia Y, Busuttil RW, Kupiec-Weglinski JW, Lu L, Wang X, Zhai Y. Glycogen synthase kinase 3β promotes liver innate immune activation by restraining AMP-activated protein kinase activation. J Hepatol 2018; 69:99-109. [PMID: 29452207 PMCID: PMC6291010 DOI: 10.1016/j.jhep.2018.01.036] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 01/08/2018] [Accepted: 01/30/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND & AIMS Glycogen synthase kinase 3β (Gsk3β [Gsk3b]) is a ubiquitously expressed kinase with distinctive functions in different types of cells. Although its roles in regulating innate immune activation and ischaemia and reperfusion injuries (IRIs) have been well documented, the underlying mechanisms remain ambiguous, in part because of the lack of cell-specific tools in vivo. METHODS We created a myeloid-specific Gsk3b knockout (KO) strain to study the function of Gsk3β in macrophages in a murine liver partial warm ischaemia model. RESULTS Compared with controls, myeloid Gsk3b KO mice were protected from IRI, with diminished proinflammatory but enhanced anti-inflammatory immune responses in livers. In bone marrow-derived macrophages, Gsk3β deficiency resulted in an early reduction of Tnf gene transcription but sustained increase of Il10 gene transcription on Toll-like receptor 4 stimulation in vitro. These effects were associated with enhanced AMP-activated protein kinase (AMPK) activation, which led to an accelerated and higher level of induction of the novel innate immune negative regulator small heterodimer partner (SHP [Nr0b2]). The regulatory function of Gsk3β on AMPK activation and SHP induction was confirmed in wild-type bone marrow-derived macrophages with a Gsk3 inhibitor. Furthermore, we found that this immune regulatory mechanism was independent of Gsk3β Ser9 phosphorylation and the phosphoinositide 3-kinase-Akt signalling pathway. In vivo, myeloid Gsk3β deficiency facilitated SHP upregulation by ischaemia-reperfusion in liver macrophages. Treatment of Gsk3b KO mice with either AMPK inhibitor or SHP small interfering RNA before the onset of liver ischaemia restored liver proinflammatory immune activation and IRI in these otherwise protected hosts. Additionally, pharmacological activation of AMPK protected wild-type mice from liver IRI, with reduced proinflammatory immune activation. Inhibition of the AMPK-SHP pathway by liver ischaemia was demonstrated in tumour resection patients. CONCLUSIONS Gsk3β promotes innate proinflammatory immune activation by restraining AMPK activation. LAY SUMMARY Glycogen synthase kinase 3β promotes macrophage inflammatory activation by inhibiting the immune regulatory signalling of AMP-activated protein kinase and the induction of small heterodimer partner. Therefore, therapeutic targeting of glycogen synthase kinase 3β enhances innate immune regulation and protects liver from ischaemia and reperfusion injury.
Collapse
Affiliation(s)
- Haoming Zhou
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA; Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Han Wang
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA; Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Ming Ni
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA; Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Shi Yue
- Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yongxiang Xia
- Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Ronald W Busuttil
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| | - Jerzy W Kupiec-Weglinski
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| | - Ling Lu
- Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xuehao Wang
- Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| | - Yuan Zhai
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA.
| |
Collapse
|
197
|
Hsu WL, Lin YC, Jeng JR, Chang HY, Chou TC. Baicalein Ameliorates Pulmonary Arterial Hypertension Caused by Monocrotaline through Downregulation of ET-1 and ETAR in Pneumonectomized Rats. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2018; 46:769-783. [DOI: 10.1142/s0192415x18500404] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Baicalein (BE) extracted from Scutellaria baicalensis Georgi is able to alleviate various cardiovascular and inflammatory diseases. However, the effects of BE on pulmonary arterial hypertension (PAH) remain unknown. Therefore, the present study aimed to examine whether BE ameliorates pneumonectomy and monocrotaline-induced PAH in rats and further investigate the underlying molecular mechanisms. Administration of BE greatly attenuated the development of PAH as evidenced by an improvement of its characteristic features, including elevation of right ventricular systolic pressure, right ventricular hypertrophy, and pulmonary vascular remodeling. Moreover, the increased protein expression of endothelin-1 (ET-1) and ETA receptor (ETAR), superoxide overproduction, and activation of Akt/ERK1/2/GSK3[Formula: see text]/[Formula: see text]-catenin pathway that occurred in the lungs of PAH rats were markedly reversed by BE treatment. Compared with the untreated PAH rats, higher expression of endothelial nitric oxide synthase (eNOS), but lower levels of inducible nitric oxide synthase and vWF were observed in BE-treated PAH rats. Collectively, treatment with BE remarkably attenuates the pathogenesis of PAH, and the protection of BE may be associated with suppressing Akt/Erk1/2/GSK3[Formula: see text]/[Formula: see text]-catenin/ET-1/ETAR signaling and preventing endothelial dysfunction. These results suggest that BE is a potential agent for treatment of PAH.
Collapse
Affiliation(s)
- Wen-Lin Hsu
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Department of Radiation Oncology, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Yu-Chieh Lin
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Jing-Ren Jeng
- Department of Cardiology, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Heng-Yuan Chang
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien, Taiwan
| | - Tz-Chong Chou
- Department of Medical Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
- China Medical University Hospital, China Medical University, Taichung, Taiwan
| |
Collapse
|
198
|
Swart PC, Russell VA, Dimatelis JJ. Maternal separation stress reduced prenatal-ethanol-induced increase in exploratory behaviour and extracellular signal-regulated kinase activity. Behav Brain Res 2018; 356:470-482. [PMID: 29908221 DOI: 10.1016/j.bbr.2018.06.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 05/22/2018] [Accepted: 06/06/2018] [Indexed: 01/26/2023]
Abstract
In an attempt to better represent the aetiology of fetal alcohol spectrum disorder (FASD) and the associated psychological deficits, prenatal-ethanol exposure was followed by maternal separation in a rat model in order to account for the effects of early-life adversities in addition to in utero alcohol exposure. Extracellular signal-regulated kinase 1/2 (ERK1/2) and glycogen synthase kinase 3-β (GSK3β) are converging points for many signalling cascades and have been implicated in models of FASD and models of early-life stress. Therefore, these kinases may also contribute to the behavioural changes observed after the combination of both developmental insults. In this study, ethanol-dams voluntarily consumed a 0.066% saccharin-sweetened 10% ethanol (EtOH) solution for 10 days prior to pregnancy and throughout gestation while control-dams had ad libitumaccess to a 0.066% saccharin (sacc) solution. Whole litters were randomly assigned to undergo maternal separation (MS) for 3 h/day from P2 to P14 while the remaining litters were left undisturbed (nMS). This resulted in 4 experimental groups: control (sacc + nMS), MS (sacc + MS), EtOH (EtOH + nMS) and EtOH + MS. Throughout development, EtOH-rats weighed less than control rats. However, subsequent maternal separation stress caused EtOH + MS-rats to weigh more than EtOH-rats. In adulthood both MS- and EtOH-rats were hyperactive but the combination produced activity levels similar to that of control rats. All treated animals (MS-, EtOH- and EtOH + MS-rats) demonstrated a negative affective state shown by increased number and duration of 22 kHz ultrasonic vocalizations compared to control rats. Prenatal-ethanol exposure increased the P-GSK3β/GSK3β ratio in the prefrontal cortex (PFC) and maternal separation decreased the P-GSK3β/GSK3β ratio in the dorsal hippocampus (DH) of adult rats. However, maternal separation stress decreased the effect of prenatal-ethanol exposure on the P-ERK/ERK ratio in the PFC and DH and reduced prenatal-ethanol-induced hyperactivity. Therefore, indicating a significant interaction between prenatal-ethanol exposure and early-life stress on behaviour and the brain and may implicate P-ERK1/2 signalling in exploratory behaviour.
Collapse
Affiliation(s)
- Patricia C Swart
- University of Cape Town, Faculty of Health Sciences, Department of Human Biology, Observatory, 7925 South Africa.
| | - Vivienne A Russell
- University of Cape Town, Faculty of Health Sciences, Department of Human Biology, Observatory, 7925 South Africa.
| | - Jacqueline J Dimatelis
- University of Cape Town, Faculty of Health Sciences, Department of Human Biology, Observatory, 7925 South Africa.
| |
Collapse
|
199
|
Schulz L, Pries R, Lanka AS, Drenckhan M, Rades D, Wollenberg B. Inhibition of GSK3α/β impairs the progression of HNSCC. Oncotarget 2018; 9:27630-27644. [PMID: 29963225 PMCID: PMC6021246 DOI: 10.18632/oncotarget.25250] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 04/06/2018] [Indexed: 12/19/2022] Open
Abstract
Background Head and neck squamous cell cancer (HNSCC) is one of the most common tumors worldwide and there is an enormous need for innovative therapy approaches. Several recent studies suggest tumor entity specific roles of glycogen synthase kinase 3 (GSK3) in different human cancers, acting as tumor suppressor or as tumor promoter. Here we describe the role of GSK3 with respect to different parameters within HNSCC progression. Methods Base line expression and activity profiles of p-GSK3α/β (Ser21/9) and p-GSK3α/β (Tyr279/216) were analyzed by immunohistochemistry and western blotting. Four different permanent HNSCC cell lines were exposed to the potent GSK3α/β inhibitor SB 216763. Cell viability was controlled via the MTT test. Cell migration was quantified with the Real Time Cell Analyzer (RCTA) xCELLigence. Regulation of the epithelial-mesenchymal transition (EMT) was measured with the Human Epithelial to Mesenchymal Transition (EMT) RT2 Profiler™ PCR Array and scratch assays. Taqman probes were used to detect the specific gene expression profiles of inflammatory cytokines Interleukin IL1β, IL6, IL8, IL10, TNFα and IFNβ. Results Exposure of permanent HNSCC cell lines to the specific GSK3α/β inhibitor SB 216763 leads to significant growth inhibition, inhibition of migration and decreased levels of active GSK3α/β in a dose dependent manner.Exposure of HNSCC lines to SB 216763 also resulted in a markable shift of EMT markers and functional EMT dysregulation. Functionally GSK3 differentially mediates the expression of TLR4- and TLR3-induced inflammatory cytokines in HNSCC, whereas no effect of SB 216763 on the NFkB activity was noticed. Conclusion GSK3α/β plays a crucial role in a variety of regulatory networks for HNSCC cancer progression as it drives proliferation or migration and thus GSK3 could serve as an interesting target for clinical drug development.
Collapse
Affiliation(s)
- Lisa Schulz
- Department of Otorhinolaryngology, University of Luebeck, Luebeck 23538, Germany
| | - Ralph Pries
- Department of Otorhinolaryngology, University of Luebeck, Luebeck 23538, Germany
| | - Aruna Sree Lanka
- Department of Otorhinolaryngology, University of Luebeck, Luebeck 23538, Germany
| | - Maren Drenckhan
- Department of Otorhinolaryngology, University of Luebeck, Luebeck 23538, Germany.,Department of Radiation Oncology, University of Luebeck, Luebeck 23538, Germany
| | - Dirk Rades
- Department of Radiation Oncology, University of Luebeck, Luebeck 23538, Germany
| | - Barbara Wollenberg
- Department of Otorhinolaryngology, University of Luebeck, Luebeck 23538, Germany
| |
Collapse
|
200
|
Cytokeratin 19 (KRT19) has a Role in the Reprogramming of Cancer Stem Cell-Like Cells to Less Aggressive and More Drug-Sensitive Cells. Int J Mol Sci 2018; 19:ijms19051423. [PMID: 29747452 PMCID: PMC5983664 DOI: 10.3390/ijms19051423] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 05/04/2018] [Accepted: 05/07/2018] [Indexed: 02/08/2023] Open
Abstract
Cytokeratin 19 (KRT19) is a cytoplasmic intermediate filament protein, which is responsible for structural rigidity and multipurpose scaffolds. In several cancers, KRT19 is overexpressed and may play a crucial role in tumorigenic transformation. In our previous study, we revealed the role of KRT19 as signaling component which mediated Wnt/NOTCH crosstalk through NUMB transcription in breast cancer. Here, we investigated the function of KRT19 in cancer reprogramming and drug resistance in breast cancer cells. We found that expression of KRT19 was attenuated in several patients-derived breast cancer tissues and patients with a low expression of KRT19 were significantly correlated with poor prognosis in breast cancer patients. Consistently, highly aggressive and drug-resistant breast cancer patient-derived cancer stem cell-like cells (konkuk university-cancer stem cell-like cell (KU-CSLCs)) displayed higher expression of cancer stem cell (CSC) markers, including ALDH1, CXCR4, and CD133, but a much lower expression of KRT19 than that is seen in highly aggressive triple negative breast cancer MDA-MB231 cells. Moreover, we revealed that the knockdown of KRT19 in MDA-MB231 cells led to an enhancement of cancer properties, such as cell proliferation, sphere formation, migration, and drug resistance, while the overexpression of KRT19 in KU-CSLCs resulted in the significant attenuation of cancer properties. KRT19 regulated cancer stem cell reprogramming by modulating the expression of cancer stem cell markers (ALDH1, CXCR4, and CD133), as well as the phosphorylation of Src and GSK3β (Tyr216). Therefore, our data may imply that the modulation of KRT19 expression could be involved in cancer stem cell reprogramming and drug sensitivity, which might have clinical implications for cancer or cancer stem cell treatment.
Collapse
|