1,060 results on '"Protein kinase C (PKC)"'
Search Results
2. Nonalcoholic Fatty Liver Disease and Staging of Hepatic Fibrosis
- Author
-
Engin, Atilla, Crusio, Wim E., Series Editor, Dong, Haidong, Series Editor, Radeke, Heinfried H., Series Editor, Rezaei, Nima, Series Editor, Steinlein, Ortrud, Series Editor, Xiao, Junjie, Series Editor, Rosenhouse-Dantsker, Avia, Editorial Board Member, ENGIN, Ayse Basak, editor, and ENGIN, Atilla, editor
- Published
- 2024
- Full Text
- View/download PDF
3. Protein Kinase C (PKC) in Neurological Health: Implications for Alzheimer's Disease and Chronic Alcohol Consumption.
- Author
-
Singh, Nishtha, Nandy, Shouvik Kumar, Jyoti, Anupam, Saxena, Juhi, Sharma, Aditi, Siddiqui, Arif Jamal, and Sharma, Lalit
- Subjects
- *
PROTEIN kinase C , *ALZHEIMER'S disease , *ALCOHOL drinking , *CHRONIC diseases , *ALCOHOLISM - Abstract
Protein kinase C (PKC) is a diverse enzyme family crucial for cell signalling in various organs. Its dysregulation is linked to numerous diseases, including cancer, cardiovascular disorders, and neurological problems. In the brain, PKC plays pivotal roles in synaptic plasticity, learning, memory, and neuronal survival. Specifically, PKC's involvement in Alzheimer's Disease (AD) pathogenesis is of significant interest. The dysregulation of PKC signalling has been linked to neurological disorders, including AD. This review elucidates PKC's pivotal role in neurological health, particularly its implications in AD pathogenesis and chronic alcohol addiction. AD, characterised by neurodegeneration, implicates PKC dysregulation in synaptic dysfunction and cognitive decline. Conversely, chronic alcohol consumption elicits neural adaptations intertwined with PKC signalling, exacerbating addictive behaviours. By unravelling PKC's involvement in these afflictions, potential therapeutic avenues emerge, offering promise for ameliorating their debilitating effects. This review navigates the complex interplay between PKC, AD pathology, and alcohol addiction, illuminating pathways for future neurotherapeutic interventions. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
4. Regulation of microtubule nucleation in mouse bone marrow-derived mast cells by ARF GTPase-activating protein GIT2.
- Author
-
Sulimenko, Vadym, Sládková, Vladimira, Sulimenko, Tetyana, Dráberová, Eduarda, Vosecká, Věra, Dráberová, Lubica, Skalli, Omar, and Dráber, Pavel
- Subjects
GTPASE-activating protein ,MAST cells ,MICROTUBULES ,SECRETORY granules ,PROTEIN kinase C - Abstract
Aggregation of high-affinity IgE receptors (FceRIs) on granulated mast cells triggers signaling pathways leading to a calcium response and release of inflammatory mediators from secretory granules. While microtubules play a role in the degranulation process, the complex molecular mechanisms regulating microtubule remodeling in activated mast cells are only partially understood. Here, we demonstrate that the activation of bone marrow mast cells induced by FceRI aggregation increases centrosomal microtubule nucleation, with G protein-coupled receptor kinase-interacting protein 2 (GIT2) playing a vital role in this process. Both endogenous and exogenous GIT2 were associated with centrosomes and γ-tubulin complex proteins. Depletion of GIT2 enhanced centrosomal microtubule nucleation, and phenotypic rescue experiments revealed that GIT2, unlike GIT1, acts as a negative regulator of microtubule nucleation in mast cells. GIT2 also participated in the regulation of antigen-induced degranulation and chemotaxis. Further experiments showed that phosphorylation affected the centrosomal localization of GIT2 and that during antigen-induced activation, GIT2 was phosphorylated by conventional protein kinase C, which promoted microtubule nucleation. We propose that GIT2 is a novel regulator of microtubule organization in activated mast cells by modulating centrosomal microtubule nucleation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
5. Pharmacological study on the enhancing effects of U46619 on guinea pig urinary bladder smooth muscle contraction induced by acetylcholine and α,β-methylene ATP and the possible involvement of protein kinase C
- Author
-
Guanghan Ou, Akane Komura, Misaki Hojo, Rina Kato, Masahiro Ikeda, Miki Fujisawa, Keyue Xu, Kento Yoshioka, Keisuke Obara, and Yoshio Tanaka
- Subjects
U46619 (a prostanoid TP receptor agonist) ,Urinary bladder smooth muscle ,Protein kinase C (PKC) ,α,β-methylene ATP ,Acetylcholine ,Therapeutics. Pharmacology ,RM1-950 - Abstract
We examined whether U46619 (a prostanoid TP receptor agonist) could enhance the contractions of guinea pig urinary bladder smooth muscle (UBSM) in response to acetylcholine (ACh) and an ATP analog (α,β-methylene ATP (αβ-MeATP)) through stimulation of the UBSM TP receptor and whether protein kinase C (PKC) is involved. U46619 (10−7 M) markedly enhanced UBSM contractions induced by electrical field stimulation and ACh/αβ-MeATP (3 × 10−6 M each), the potentiation of which was completely suppressed by SQ 29,548 (a TP receptor antagonist, 6 × 10−7 M). PKC inhibitors did not attenuate the ACh-induced contractions enhanced by U46619 although they partly suppressed the U46619-enhanced, αβ-MeATP-induced contractions. While phorbol 12-myristate 13-acetate (PMA, a PKC activator, 10−6 M) did not enhance ACh-induced contractions, it enhanced αβ-MeATP-induced contractions, an effect that was completely suppressed by PKC inhibitors. αβ-MeATP-induced contractions, both with and without U46619 enhancement, were strongly inhibited by diltiazem. U46619/PMA enhanced 50 mM KCl-induced contractions, the potentiation of which was partly/completely attenuated by PKC inhibitors. These findings suggest that U46619 potentiates parasympathetic nerve-associated UBSM contractions by stimulating UBSM TP receptors. PKC-increased Ca2+ influx through voltage-dependent Ca2+ channels may partially play a role in purinergic receptor-mediated UBSM contractions enhanced by TP receptor stimulation.
- Published
- 2023
- Full Text
- View/download PDF
6. Review of Neuraxial Agents Producing Analgesia
- Author
-
Dias, Elayne Vieira, Sorkin, Linda S., Yaksh, Tony L., Yaksh, Tony, editor, and Hayek, Salim, editor
- Published
- 2023
- Full Text
- View/download PDF
7. Regulation of microtubule nucleation in mouse bone marrow-derived mast cells by ARF GTPase-activating protein GIT2
- Author
-
Vadym Sulimenko, Vladimíra Sládková, Tetyana Sulimenko, Eduarda Dráberová, Věra Vosecká, Lubica Dráberová, Omar Skalli, and Pavel Dráber
- Subjects
mast cells ,centrosome ,G protein-coupled receptor kinase-interacting protein 2 (GIT2) ,microtubule nucleation ,protein kinase C (PKC) ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Aggregation of high-affinity IgE receptors (FcϵRIs) on granulated mast cells triggers signaling pathways leading to a calcium response and release of inflammatory mediators from secretory granules. While microtubules play a role in the degranulation process, the complex molecular mechanisms regulating microtubule remodeling in activated mast cells are only partially understood. Here, we demonstrate that the activation of bone marrow mast cells induced by FcϵRI aggregation increases centrosomal microtubule nucleation, with G protein-coupled receptor kinase-interacting protein 2 (GIT2) playing a vital role in this process. Both endogenous and exogenous GIT2 were associated with centrosomes and γ-tubulin complex proteins. Depletion of GIT2 enhanced centrosomal microtubule nucleation, and phenotypic rescue experiments revealed that GIT2, unlike GIT1, acts as a negative regulator of microtubule nucleation in mast cells. GIT2 also participated in the regulation of antigen-induced degranulation and chemotaxis. Further experiments showed that phosphorylation affected the centrosomal localization of GIT2 and that during antigen-induced activation, GIT2 was phosphorylated by conventional protein kinase C, which promoted microtubule nucleation. We propose that GIT2 is a novel regulator of microtubule organization in activated mast cells by modulating centrosomal microtubule nucleation.
- Published
- 2024
- Full Text
- View/download PDF
8. An Update on Protein Kinases as Therapeutic Targets—Part I: Protein Kinase C Activation and Its Role in Cancer and Cardiovascular Diseases.
- Author
-
Silnitsky, Shmuel, Rubin, Samuel J. S., Zerihun, Mulate, and Qvit, Nir
- Subjects
- *
PROTEIN kinases , *DRUG target , *PROTEIN kinase inhibitors , *CARDIOVASCULAR diseases , *ALLOSTERIC regulation - Abstract
Protein kinases are one of the most significant drug targets in the human proteome, historically harnessed for the treatment of cancer, cardiovascular disease, and a growing number of other conditions, including autoimmune and inflammatory processes. Since the approval of the first kinase inhibitors in the late 1990s and early 2000s, the field has grown exponentially, comprising 98 approved therapeutics to date, 37 of which were approved between 2016 and 2021. While many of these small-molecule protein kinase inhibitors that interact orthosterically with the protein kinase ATP binding pocket have been massively successful for oncological indications, their poor selectively for protein kinase isozymes have limited them due to toxicities in their application to other disease spaces. Thus, recent attention has turned to the use of alternative allosteric binding mechanisms and improved drug platforms such as modified peptides to design protein kinase modulators with enhanced selectivity and other pharmacological properties. Herein we review the role of different protein kinase C (PKC) isoforms in cancer and cardiovascular disease, with particular attention to PKC-family inhibitors. We discuss translational examples and carefully consider the advantages and limitations of each compound (Part I). We also discuss the recent advances in the field of protein kinase modulators, leverage molecular docking to model inhibitor–kinase interactions, and propose mechanisms of action that will aid in the design of next-generation protein kinase modulators (Part II). [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
9. Features and mechanisms of propofol-induced protein kinase C (PKC) translocation and activation in living cells.
- Author
-
Soma Noguchi, Taketoshi Kajimoto, Takuya Kumamoto, Masashi Shingai, Soshi Narasaki, Tomoaki Urabe, Serika Imamura, Kana Harada, Izumi Hide, Sigeru Tanaka, Yuhki Yanase, Shun-Ichi Nakamura, Tsutsumi, Yasuo M., and Norio Sakai
- Subjects
PROTEIN kinase C ,NUCLEAR membranes ,GREEN fluorescent protein ,GOLGI apparatus ,HELA cells ,CELL membranes - Abstract
Background and purpose: In this study, we aimed to elucidate the action mechanisms of propofol, particularly those underlying propofol-induced protein kinase C (PKC) translocation. Experimental approach: Various PKCs fused with green fluorescent protein (PKCGFP) or other GFP-fused proteins were expressed in HeLa cells, and their propofol-induced dynamics were observed using confocal laser scanning microscopy. Propofol-induced PKC activation in cells was estimated using the C kinase activity receptor (CKAR), an indicator of intracellular PKC activation. We also examined PKC translocation using isomers and derivatives of propofol to identify the crucial structural motifs involved in this process. Key results: Propofol persistently translocated PKCα conventional PKCs and PKCδ from novel PKCs (nPKCs) to the plasma membrane (PM). Propofol translocated PKCδ and PKCη of nPKCs to the Golgi apparatus and endoplasmic reticulum, respectively. Propofol also induced the nuclear translocation of PKCζ of atypical PKCs or proteins other than PKCs, such that the protein concentration inside and outside the nucleus became uniform. CKAR analysis revealed that propofol activated PKC in the PM and Golgi apparatus. Moreover, tests using isomers and derivatives of propofol predicted that the structural motifs important for the induction of PKC and nuclear translocation are different. Conclusion and implications: Propofol induced the subtype-specific intracellular translocation of PKCs and activated PKCs. Additionally, propofol induced the nuclear translocation of PKCs and other proteins, probably by altering the permeability of the nuclear envelope. Interestingly, propofol-induced PKC and nuclear translocation may occur via different mechanisms. Our findings provide insights into the action mechanisms of propofol. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
10. Neuroproteomic mapping of kinases and their substrates downstream of acetylcholine: finding and implications.
- Author
-
Yamahashi, Yukie, Tsuboi, Daisuke, Funahashi, Yasuhiro, and Kaibuchi, Kozo
- Abstract
Since the emergence of the cholinergic hypothesis of Alzheimer's disease (AD), acetylcholine has been viewed as a mediator of learning and memory. Donepezil improves AD-associated learning deficits and memory loss by recovering brain acetylcholine levels. However, it is associated with side effects due to global activation of acetylcholine receptors. Muscarinic acetylcholine receptor M1 (M1R), a key mediator of learning and memory, has been an alternative target. The importance of targeting a specific pathway downstream of M1R has recently been recognized. Elucidating signaling pathways beyond M1R that lead to learning and memory holds important clues for AD therapeutic strategies. This review first summarizes the role of acetylcholine in aversive learning, one of the outputs used for preliminary AD drug screening. It then describes the phosphoproteomic approach focused on identifying acetylcholine intracellular signaling pathways leading to aversive learning. Finally, the intracellular mechanism of donepezil and its effect on learning and memory is discussed. The elucidation of signaling pathways beyond M1R by phosphoproteomic approach offers a platform for understanding the intracellular mechanism of AD drugs and for developing AD therapeutic strategies. Clarifying the molecular mechanism that links the identified acetylcholine signaling to AD pathophysiology will advance the development of AD therapeutic strategies. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
11. The Antioxidative Effects of Flavones in Hypertensive Disease.
- Author
-
Haynes, Alexandria Porcia, Desta, Selam, Ahmad, Taseer, Neikirk, Kit, Hinton, Antentor, Bloodworth, Nathaniel, and Kirabo, Annet
- Subjects
FLAVONES ,THERAPEUTIC communities ,NADPH oxidase ,FLAVONOIDS ,SODIUM channels - Abstract
Hypertension is the leading remediable risk factor for cardiovascular morbidity and mortality in the United States. Excess dietary salt consumption, which is a catalyst of hypertension, initiates an inflammatory cascade via activation of antigen-presenting cells (APCs). This pro-inflammatory response is driven primarily by sodium ions (Na
+ ) transporting into APCs by the epithelial sodium channel (ENaC) and subsequent NADPH oxidase activation, leading to high levels of oxidative stress. Oxidative stress, a well-known catalyst for hypertension-related illness development, disturbs redox homeostasis, which ultimately promotes lipid peroxidation, isolevuglandin production and an inflammatory response. Natural medicinal compounds derived from organic materials that are characterized by their anti-inflammatory, anti-oxidative, and anti-mutagenic properties have recently gained traction amongst the pharmacology community due to their therapeutic effects. Flavonoids, a natural phenolic compound, have these therapeutic benefits and can potentially serve as anti-hypertensives. Flavones are a type of flavonoid that have increased anti-inflammatory effects that may allow them to act as therapeutic agents for hypertension, including diosmetin, which is able to induce significant arterial vasodilation in several different animal models. This review will focus on the activity of flavones to illuminate potential preventative and potential therapeutic mechanisms against hypertension. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
12. Protein Kinase C (PKC) in Neurological Health: Implications for Alzheimer’s Disease and Chronic Alcohol Consumption
- Author
-
Nishtha Singh, Shouvik Kumar Nandy, Anupam Jyoti, Juhi Saxena, Aditi Sharma, Arif Jamal Siddiqui, and Lalit Sharma
- Subjects
protein kinase C (PKC) ,brain ,AD ,synaptic plasticity ,neurotransmitter regulation ,neuronal signalling ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Protein kinase C (PKC) is a diverse enzyme family crucial for cell signalling in various organs. Its dysregulation is linked to numerous diseases, including cancer, cardiovascular disorders, and neurological problems. In the brain, PKC plays pivotal roles in synaptic plasticity, learning, memory, and neuronal survival. Specifically, PKC’s involvement in Alzheimer’s Disease (AD) pathogenesis is of significant interest. The dysregulation of PKC signalling has been linked to neurological disorders, including AD. This review elucidates PKC’s pivotal role in neurological health, particularly its implications in AD pathogenesis and chronic alcohol addiction. AD, characterised by neurodegeneration, implicates PKC dysregulation in synaptic dysfunction and cognitive decline. Conversely, chronic alcohol consumption elicits neural adaptations intertwined with PKC signalling, exacerbating addictive behaviours. By unravelling PKC’s involvement in these afflictions, potential therapeutic avenues emerge, offering promise for ameliorating their debilitating effects. This review navigates the complex interplay between PKC, AD pathology, and alcohol addiction, illuminating pathways for future neurotherapeutic interventions.
- Published
- 2024
- Full Text
- View/download PDF
13. Elucidating the role of missense SNP of protein kinase C epsilon in HCV-induced hepatocellular carcinoma
- Author
-
Areeba Rehman, Maria Shabbir, Yasmin Badshah, Khushbukhat Khan, Janeen H. Trembley, Naeem Mahmood Ashraf, Tayyaba Afsar, Ali Almajwal, Nawaf W. Alruwaili, Ali Alshamari, Tariq Nahar Alanezi, and Suhail Razak
- Subjects
Protein kinase C (PKC) ,Hepatocellular carcinoma (HCC) ,Single nucleotide polymorphism (SNP) ,Hepatitis C virus (HCV) ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background The protein kinase C (PKC) family of serine/threonine kinases contains more than ten isozymes that are involved in multiple signaling pathways, including cell cycle regulation and carcinogenesis. The PKCε isozyme is an oncogene known to be upregulated in various signaling pathways involved in hepatitis C virus (HCV)-induced hepatocellular carcinoma (HCC). However, there is no known association of missense SNPs in PKCε with this disease, which can be a potential biomarker for early diagnosis and treatment. This research reveals a novel missense SNP in PKCε that is associated with HCV-induced HCC in the Pakistani population. Methods The PKCε SNP with amino acid substitution of E14K was chosen for wet lab analysis. Tetra ARMS-PCR was employed for the identification of high-risk SNP in PKCε of HCV-induced HCC patients. Liver function testing was also performed for comparison between the liver condition of the HCC patient and control group, and the viral load of HCC patient samples was evaluated to determine any alteration in the viral infectivity between different genotypes of the selected high-risk PKCε variant SNP. Results Frequency distribution of the homozygous GG genotype was found to be highest among HCV-induced HCC patients and was also found to be significantly associated with disease development and progression. The p values of comparative data obtained for the other two genotypes, heterozygous AG and homozygous AA, of the SNP also showed the significance of the data for these alleles. Still, their odds ratio and relative risk analysis did not indicate their association with HCV-induced HCC. Conclusion The distribution of a genotype GG of PKCε has been found in HCV- induced HCC patients. Therefore, these PKCε SNP have the potential to be biomarkers for HCV-induced HCC. Further investigation using a larger sample size would provide additional insight into these initial data and open a new avenue for a better prognosis of this disease.
- Published
- 2023
- Full Text
- View/download PDF
14. Evidence for Angiotensin II as a Naturally Existing Suppressor for the Guanylyl Cyclase A Receptor and Cyclic GMP Generation.
- Author
-
Ma, Xiao, Iyer, Seethalakshmi R., Ma, Xiaoyu, Reginauld, Shawn H., Chen, Yang, Pan, Shuchong, Zheng, Ye, Moroni, Dante G., Yu, Yue, Zhang, Lianwen, Cannone, Valentina, Chen, Horng H., Ferrario, Carlos M., Sangaralingham, S. Jeson, and Burnett Jr., John C.
- Subjects
- *
GUANYLATE cyclase , *CYCLIC guanylic acid , *NATRIURETIC peptides , *PROTEIN kinase C , *PEPTIDES , *ANGIOTENSIN II , *BRAIN natriuretic factor - Abstract
The natriuretic peptide system (NPS) and renin-angiotensin-aldosterone system (RAAS) function oppositely at multiple levels. While it has long been suspected that angiotensin II (ANGII) may directly suppress NPS activity, no clear evidence to date supports this notion. This study was designed to systematically investigate ANGII–NPS interaction in humans, in vivo, and in vitro. Circulating atrial, b-type, and c-type natriuretic peptides (ANP, BNP, CNP), cyclic guanosine monophosphate (cGMP), and ANGII were simultaneously investigated in 128 human subjects. Prompted hypothesis was validated in vivo to determine the influence of ANGII on ANP actions. The underlying mechanisms were further explored via in vitro approaches. In humans, ANGII demonstrated an inverse relationship with ANP, BNP, and cGMP. In regression models predicting cGMP, adding ANGII levels and the interaction term between ANGII and natriuretic peptides increased the predictive accuracy of the base models constructed with either ANP or BNP, but not CNP. Importantly, stratified correlation analysis further revealed a positive association between cGMP and ANP or BNP only in subjects with low, but not high, ANGII levels. In rats, co-infusion of ANGII even at a physiological dose attenuated cGMP generation mediated by ANP infusion. In vitro, we found the suppressive effect of ANGII on ANP-stimulated cGMP requires the presence of ANGII type-1 (AT1) receptor and mechanistically involves protein kinase C (PKC), as this suppression can be substantially rescued by either valsartan (AT1 blocker) or Go6983 (PKC inhibitor). Using surface plasmon resonance (SPR), we showed ANGII has low binding affinity to the guanylyl cyclase A (GC-A) receptor compared to ANP or BNP. Our study reveals ANGII is a natural suppressor for the cGMP-generating action of GC-A via AT1/PKC dependent manner and highlights the importance of dual-targeting RAAS and NPS in maximizing beneficial properties of natriuretic peptides in cardiovascular protection. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
15. Extracellular Histone-Induced Protein Kinase C Alpha Activation and Troponin Phosphorylation Is a Potential Mechanism of Cardiac Contractility Depression in Sepsis.
- Author
-
Abrams, Simon T., Alhamdi, Yasir, Zi, Min, Guo, Fengmei, Du, Min, Wang, Guozheng, Cartwright, Elizabeth J., and Toh, Cheng-Hock
- Subjects
- *
PROTEIN kinase C , *SEPSIS , *CALCIUM-dependent protein kinase , *TROPONIN , *TROPONIN I , *INTRACELLULAR calcium - Abstract
Reduction in cardiac contractility is common in severe sepsis. However, the pathological mechanism is still not fully understood. Recently it has been found that circulating histones released after extensive immune cell death play important roles in multiple organ injury and disfunction, particularly in cardiomyocyte injury and contractility reduction. How extracellular histones cause cardiac contractility depression is still not fully clear. In this work, using cultured cardiomyocytes and a histone infusion mouse model, we demonstrate that clinically relevant histone concentrations cause significant increases in intracellular calcium concentrations with subsequent activation and enriched localization of calcium-dependent protein kinase C (PKC) α and βII into the myofilament fraction of cardiomyocytes in vitro and in vivo. Furthermore, histones induced dose-dependent phosphorylation of cardiac troponin I (cTnI) at the PKC-regulated phosphorylation residues (S43 and T144) in cultured cardiomyocytes, which was also confirmed in murine cardiomyocytes following intravenous histone injection. Specific inhibitors against PKCα and PKCβII revealed that histone-induced cTnI phosphorylation was mainly mediated by PKCα activation, but not PKCβII. Blocking PKCα also significantly abrogated histone-induced deterioration in peak shortening, duration and the velocity of shortening, and re-lengthening of cardiomyocyte contractility. These in vitro and in vivo findings collectively indicate a potential mechanism of histone-induced cardiomyocyte dysfunction driven by PKCα activation with subsequent enhanced phosphorylation of cTnI. These findings also indicate a potential mechanism of clinical cardiac dysfunction in sepsis and other critical illnesses with high levels of circulating histones, which holds the potential translational benefit to these patients by targeting circulating histones and downstream pathways. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
16. Elucidating the role of missense SNP of protein kinase C epsilon in HCV-induced hepatocellular carcinoma.
- Author
-
Rehman, Areeba, Shabbir, Maria, Badshah, Yasmin, Khan, Khushbukhat, Trembley, Janeen H., Ashraf, Naeem Mahmood, Afsar, Tayyaba, Almajwal, Ali, Alruwaili, Nawaf W., Alshamari, Ali, Alanezi, Tariq Nahar, and Razak, Suhail
- Subjects
- *
PROTEIN kinase C , *HEPATOCELLULAR carcinoma , *SINGLE nucleotide polymorphisms , *CELL cycle regulation , *PAKISTANIS - Abstract
Background: The protein kinase C (PKC) family of serine/threonine kinases contains more than ten isozymes that are involved in multiple signaling pathways, including cell cycle regulation and carcinogenesis. The PKCε isozyme is an oncogene known to be upregulated in various signaling pathways involved in hepatitis C virus (HCV)-induced hepatocellular carcinoma (HCC). However, there is no known association of missense SNPs in PKCε with this disease, which can be a potential biomarker for early diagnosis and treatment. This research reveals a novel missense SNP in PKCε that is associated with HCV-induced HCC in the Pakistani population. Methods: The PKCε SNP with amino acid substitution of E14K was chosen for wet lab analysis. Tetra ARMS-PCR was employed for the identification of high-risk SNP in PKCε of HCV-induced HCC patients. Liver function testing was also performed for comparison between the liver condition of the HCC patient and control group, and the viral load of HCC patient samples was evaluated to determine any alteration in the viral infectivity between different genotypes of the selected high-risk PKCε variant SNP. Results: Frequency distribution of the homozygous GG genotype was found to be highest among HCV-induced HCC patients and was also found to be significantly associated with disease development and progression. The p values of comparative data obtained for the other two genotypes, heterozygous AG and homozygous AA, of the SNP also showed the significance of the data for these alleles. Still, their odds ratio and relative risk analysis did not indicate their association with HCV-induced HCC. Conclusion: The distribution of a genotype GG of PKCε has been found in HCV- induced HCC patients. Therefore, these PKCε SNP have the potential to be biomarkers for HCV-induced HCC. Further investigation using a larger sample size would provide additional insight into these initial data and open a new avenue for a better prognosis of this disease. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
17. Positioning-dependent bidirectional NELL2 signaling in the brain.
- Author
-
Byung Ju Lee and Jin Kwon Jeong
- Published
- 2022
- Full Text
- View/download PDF
18. Pathogenesis of Microvascular Complications
- Author
-
Khamaisi, Mogher, King, George L., Park, Kyoungmin, Li, Qian, Lenzi, Andrea, Series Editor, Jannini, Emmanuele A., Series Editor, Bonora, Enzo, editor, and DeFronzo, Ralph A., editor
- Published
- 2020
- Full Text
- View/download PDF
19. Anti-diabetic activity of extract from Morus nigra L. twigs through activation of AMPK/PKC pathway in mice
- Author
-
Ping Tang, Xinzhou Yang, Huijian Chen, Ting Zhang, Hui Tang, and Kejian Pang
- Subjects
Morus nigra L. ,Type 2 diabetes mellitus ,Glucose transporter 4 (GLUT4) ,Protein kinase C (PKC) ,AMP-activated protein kinase (AMPK) ,Nutrition. Foods and food supply ,TX341-641 - Abstract
Morus nigra L. is a traditional ethnomedicinal plants, which is used for the treatment of atherosclerosis, diabetes, hyperlipidemia, and hypertension. The aim of this study was to investigate the antidiabetic activity and potential mechanisms of ethyl acetate extract of Morus nigra L. twigs (MNT-EA). In L6 cells, MNT-EA significantly upregulated glucose uptake, and promoted GLUT4 translocation and expression through AMPK and PKC signaling pathway. In vivo, diabetic mice treated with MNT-EA for 4 weeks improved hyperglycemia, lipid metabolism disorders and insulin resistance. According to histopathological examination, MNT-EA attenuated hepatic steatosis, adipocyte hypertrophy, and islet lesions in diabetic mice. In addition, MNT-EA upregulated p-AMPK, p-PKC, and GLUT4 expression in liver, adipose, and skeletal muscle tissues, thereby enhancing glucose uptake and improving diabetic symptoms. It is the first time to demonstrate that MNT-EA exerted anti-diabetic effects through activation of AMPK/PKC/GLUT4 signaling pathway, providing a basis for the development of new anti-diabetic drugs.
- Published
- 2022
- Full Text
- View/download PDF
20. Maprotiline Ameliorates High Glucose-Induced Dysfunction in Renal Glomerular Endothelial Cells.
- Author
-
Zhou, Zhihong and Liu, Shangjun
- Subjects
- *
ENDOTHELIAL cells , *KIDNEY diseases , *DIABETIC nephropathies , *PROTEIN kinases , *SUPEROXIDE dismutase - Abstract
Maprotiline is an antidepressant that has been found to cause hypoglycemia. However, the effect of maprotiline on diabetic nephropathy (DN) has not been investigated. Here, we explored the effect of maprotiline on human renal glomerular endothelial cells (HRGECs) in response to high glucose (HG) stimulation. We found that maprotiline attenuated HG-induced oxidative stress in HRGECs with decreased reactive oxygen species production and increased superoxide dismutase activity. Maprotiline repressed the HG-induced expression of cyclooxygenases 2 at both mRNA and protein levels in HRGECs. The increased thromboxane B2 level and decreased 6-keto-prostaglandin F1α level induced by HG were significantly attenuated by maprotiline treatment. Maprotiline also prevented the HG-induced increase in the permeability of HRGECs and the decrease in the zonula occludens-1 expression and downregulated HG-induced increase in the expression of protein kinase C-α (PKC-α) in HRGECs. This protective effect of maprotiline on HG-induced HRGECs dysfunction was abolished by overexpression of PKC-α. In conclusion, maprotiline displayed a protective effect on HG-challenged HRGECs, which was mediated by the regulation of PKC-α. These findings provide further evidence for the potential use of maprotiline for the treatment of DN. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
21. Selective protein kinase C inhibition switches time-dependent glucose cardiotoxicity to cardioprotection
- Author
-
Sean Brennan, Simona Esposito, Muhammad I. M. Abdelaziz, Christopher A. Martin, Samir Makwana, Mark W. Sims, Iain B. Squire, Parveen Sharma, Amy E. Chadwick, and Richard D. Rainbow
- Subjects
glucose ,cardiotoxicity ,cardioprotection ,hyperglycaemia ,protein kinase C (PKC) ,time-dependent ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Hyperglycaemia at the time of myocardial infarction has an adverse effect on prognosis irrespective of a prior diagnosis of diabetes, suggesting glucose is the damaging factor. In ex vivo models of ischaemia, we demonstrated that deleterious effects of acutely elevated glucose are PKCα/β-dependent, and providing PKCα/β are inhibited, elevated glucose confers cardioprotection. Short pre-treatments with high glucose were used to investigate time-dependent glucose cardiotoxicity, with PKCα/β inhibition investigated as a potential mechanism to reverse the toxicity. Freshly isolated non-diabetic rat cardiomyocytes were exposed to elevated glucose to investigate the time-dependence toxic effects. High glucose challenge for >7.5 min was cardiotoxic, proarrhythmic and lead to contractile failure, whilst cardiomyocytes exposed to metabolic inhibition following 5-min high glucose, displayed a time-dependent protection lasting ∼15 min. This protection was further enhanced with PKCα/β inhibition. Cardioprotection was measured as a delay in contractile failure and KATP channel activation, improved contractile and Ca2+ transient recovery and increased cell survival. Finally, the effects of pre-ischaemic treatment with high glucose in a whole-heart coronary ligation protocol, where protection was evident with PKCα/β inhibition. Selective PKCα/β inhibition enhances protection suggesting glycaemic control with PKC inhibition as a potential cardioprotective therapeutics in myocardial infarction and elective cardiac surgery.
- Published
- 2022
- Full Text
- View/download PDF
22. Antitumor activity of adiponectin in mice model of skin cancer via modulation of sulfatase-2 and MMP9 activity on HSPGs.
- Author
-
Alyoussef, Abdullah
- Subjects
- *
SKIN cancer , *ADIPONECTIN , *PROTEIN kinase C , *HEPARAN sulfate proteoglycans , *ANIMAL disease models , *MATRIX metalloproteinases - Abstract
Incidence and progression of skin cancer has progressively elevated in the last decades because of the increase in risk factors such as ultraviolet radiation. In addition, adiponectin exerts therapeutic effects against many types of tumors. Therefore, we aimed to investigate therapeutic effects of adiponectin on skin cancer and to study its effects on inflammation and tumor invasion in a mice model of skin cancer. Skin cancer was induced using 7,12-Dimethylbenz (a) anthracene (DMBA) and croton oil on the dorsal skin of mice. Skin samples were obtained for estimation of gene and protein expression of sulfatase-2, matrix metalloproteinase (MMP)9, heparan sulfate proteoglycans (HSPGs), glypican-3 (GPC3), fascin, nuclear factor (NF)κB; protein kinase C (PKC) and syndecan-1. Part of the skin is immune stained with cytokeratin. Skin cancer-induced elevation in activity of sulfatase-2 and MMP9 enzymes leading to reduction in HSPGs. All these effects were ameliorated by adiponectin. Finally, treatment with adiponectin markedly attenuated skin cancer-induced elevation in the expression of PKC, NFκB, fascin and syndecan-1. In conclusion, besides its antioxidant activities, adiponectin produced therapeutic effects against skin cancer through increasing HSPGs and inhibition of both inflammatory and tumor invasion pathways. Highlights: • Adiponectin exerts therapeutic effects against many types of tumors. • Treatment of skin cancer mice with adiponectin increases HSPGs. • Adiponectin inhibits both inflammatory and tumor invasion pathways. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
23. Role of Cortisol and Dehydroepiandrosterone on RACK1/PKC Signalling and Consequences in Immunosenescence
- Author
-
Buoso, E., Serafini, Mm., Galasso, M., Ronfani, M., Poloni, L., Lanni, C., Corsini, E., Racchi, M., Fulop, Tamas, editor, Franceschi, Claudio, editor, Hirokawa, Katsuiku, editor, and Pawelec, Graham, editor
- Published
- 2019
- Full Text
- View/download PDF
24. Deciphering the Role of PKC in Calpain-CAST System Through Formal Modeling Approach
- Author
-
Ashraf, Javaria, Ahmad, Jamil, Ul-Haq, Zaheer, Hutchison, David, Editorial Board Member, Kanade, Takeo, Editorial Board Member, Kittler, Josef, Editorial Board Member, Kleinberg, Jon M., Editorial Board Member, Mattern, Friedemann, Editorial Board Member, Mitchell, John C., Editorial Board Member, Naor, Moni, Editorial Board Member, Pandu Rangan, C., Editorial Board Member, Steffen, Bernhard, Editorial Board Member, Terzopoulos, Demetri, Editorial Board Member, Tygar, Doug, Editorial Board Member, Goos, Gerhard, Founding Editor, Hartmanis, Juris, Founding Editor, Rojas, Ignacio, editor, Valenzuela, Olga, editor, Rojas, Fernando, editor, and Ortuño, Francisco, editor
- Published
- 2019
- Full Text
- View/download PDF
25. Cardiomyocyte stromal interaction molecule 1 is a key regulator of Ca2+‐dependent kinase and phosphatase activity in the mouse heart.
- Author
-
Collins, Helen E., Anderson, Joshua C., Wende, Adam R., and Chatham, John C.
- Subjects
- *
PROTEIN kinase C , *CGMP-dependent protein kinase , *HEART , *MITOGEN-activated protein kinase phosphatases , *DILATED cardiomyopathy , *MOLECULES - Abstract
Stromal interaction molecule 1 (STIM1) is a major regulator of store‐operated calcium entry in non‐excitable cells. Recent studies have suggested that STIM1 plays a role in pathological hypertrophy; however, the physiological role of STIM1 in the heart is not well understood. We have shown that mice with a cardiomyocyte deletion of STIM1 (crSTIM1−/−) develop ER stress, mitochondrial, and metabolic abnormalities, and dilated cardiomyopathy. However, the specific signaling pathways and kinases regulated by STIM1 are largely unknown. Therefore, we used a discovery‐based kinomics approach to identify kinases differentially regulated by STIM1. Twelve‐week male control and crSTIM1−/− mice were injected with saline or phenylephrine (PE, 15 mg/kg, s.c, 15 min), and hearts obtained for analysis of the Serine/threonine kinome. Primary analysis was performed using BioNavigator 6.0 (PamGene), using scoring from the Kinexus PhosphoNET database and GeneGo network modeling, and confirmed using standard immunoblotting. Kinomics revealed significantly lower PKG and protein kinase C (PKC) signaling in the hearts of the crSTIM1−/− in comparison to control hearts, confirmed by immunoblotting for the calcium‐dependent PKC isoform PKCα and its downstream target MARCKS. Similar reductions in crSTIM1−/− hearts were found for the kinases: MEK1/2, AMPK, and PDPK1, and in the activity of the Ca2+‐dependent phosphatase, calcineurin. Electrocardiogram analysis also revealed that crSTIM1−/− mice have significantly lower HR and prolonged QT interval. In conclusion, we have shown several calcium‐dependent kinases and phosphatases are regulated by STIM1 in the adult mouse heart. This has important implications in understanding how STIM1 contributes to the regulation of cardiac physiology and pathophysiology. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
26. Muscarinic signaling regulates voltage‐gated potassium channel KCNQ2 phosphorylation in the nucleus accumbens via protein kinase C for aversive learning.
- Author
-
Faruk, Md. Omar, Tsuboi, Daisuke, Yamahashi, Yukie, Funahashi, Yasuhiro, Lin, You‐Hsin, Ahammad, Rijwan Uddin, Hossen, Emran, Amano, Mutsuki, Nishioka, Tomoki, Tzingounis, Anastasios V, Yamada, Kiyofumi, Nagai, Taku, and Kaibuchi, Kozo
- Subjects
- *
PROTEIN kinase C , *POTASSIUM channels , *MUSCARINIC receptors , *MUSCARINIC acetylcholine receptors , *NUCLEUS accumbens , *AVERSIVE stimuli , *PHOSPHORYLATION , *ELECTRIC shock - Abstract
The nucleus accumbens (NAc) plays critical roles in emotional behaviors, including aversive learning. Aversive stimuli such as an electric foot shock increase acetylcholine (ACh) in the NAc, and muscarinic signaling appears to increase neuronal excitability and aversive learning. Muscarinic signaling inhibits the voltage‐dependent potassium KCNQ current which regulates neuronal excitability, but the regulatory mechanism has not been fully elucidated. Phosphorylation of KCNQ2 at threonine 217 (T217) and its inhibitory effect on channel activity were predicted. However, whether and how muscarinic signaling phosphorylates KCNQ2 in vivo remains unclear. Here, we found that PKC directly phosphorylated KCNQ2 at T217 in vitro. Carbachol and a muscarinic M1 receptor (M1R) agonist facilitated KCNQ2 phosphorylation at T217 in NAc/striatum slices in a PKC‐dependent manner. Systemic administration of the cholinesterase inhibitor donepezil, which is commonly used to treat dementia, and electric foot shock to mice induced the phosphorylation of KCNQ2 at T217 in the NAc, whereas phosphorylation was suppressed by an M1R antagonist. Conditional deletion of Kcnq2 in the NAc enhanced electric foot shock induced aversive learning. Our findings indicate that muscarinic signaling induces the phosphorylation of KCNQ2 at T217 via PKC activation for aversive learning. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
27. Cardiomyocyte stromal interaction molecule 1 is a key regulator of Ca2+‐dependent kinase and phosphatase activity in the mouse heart
- Author
-
Helen E. Collins, Joshua C. Anderson, Adam R. Wende, and John C. Chatham
- Subjects
calcium‐dependent ,cardiomyocytes ,kinases ,protein kinase C (PKC) ,protein kinase G (PKG) ,store‐operated calcium entry (SOCE) ,Physiology ,QP1-981 - Abstract
Abstract Stromal interaction molecule 1 (STIM1) is a major regulator of store‐operated calcium entry in non‐excitable cells. Recent studies have suggested that STIM1 plays a role in pathological hypertrophy; however, the physiological role of STIM1 in the heart is not well understood. We have shown that mice with a cardiomyocyte deletion of STIM1 (crSTIM1−/−) develop ER stress, mitochondrial, and metabolic abnormalities, and dilated cardiomyopathy. However, the specific signaling pathways and kinases regulated by STIM1 are largely unknown. Therefore, we used a discovery‐based kinomics approach to identify kinases differentially regulated by STIM1. Twelve‐week male control and crSTIM1−/− mice were injected with saline or phenylephrine (PE, 15 mg/kg, s.c, 15 min), and hearts obtained for analysis of the Serine/threonine kinome. Primary analysis was performed using BioNavigator 6.0 (PamGene), using scoring from the Kinexus PhosphoNET database and GeneGo network modeling, and confirmed using standard immunoblotting. Kinomics revealed significantly lower PKG and protein kinase C (PKC) signaling in the hearts of the crSTIM1−/− in comparison to control hearts, confirmed by immunoblotting for the calcium‐dependent PKC isoform PKCα and its downstream target MARCKS. Similar reductions in crSTIM1−/− hearts were found for the kinases: MEK1/2, AMPK, and PDPK1, and in the activity of the Ca2+‐dependent phosphatase, calcineurin. Electrocardiogram analysis also revealed that crSTIM1−/− mice have significantly lower HR and prolonged QT interval. In conclusion, we have shown several calcium‐dependent kinases and phosphatases are regulated by STIM1 in the adult mouse heart. This has important implications in understanding how STIM1 contributes to the regulation of cardiac physiology and pathophysiology.
- Published
- 2022
- Full Text
- View/download PDF
28. Pathogenesis of Microvascular Complications
- Author
-
Khamaisi, Mogher, King, George L., Park, Kyoungmin, Li, Qian, Lenzi, Andrea, Series Editor, Jannini, Emmanuele A., Series Editor, Bonora, Enzo, editor, and DeFronzo, Ralph A., editor
- Published
- 2018
- Full Text
- View/download PDF
29. Possible Association Between DHEA and PKCε in Hepatic Encephalopathy Amelioration: A Pilot Study
- Author
-
Alessandro Di Cerbo, Luca Roncati, Carlotta Marini, Gianluca Carnevale, Manuela Zavatti, Rossella Avallone, and Lorenzo Corsi
- Subjects
hepatic encephalopathy ,hyperammonemia ,protein kinase C (PKC) ,DHEA ,animal model ,Veterinary medicine ,SF600-1100 - Abstract
Objective: Hepatic encephalopathy (HE) is a neuropsychiatric syndrome caused by liver failure and by an impaired neurotransmission and neurological function caused by hyperammonemia (HA). HE, in turn, decreases the phosphorylation of protein kinase C epsilon (PKCε), contributing to the impairment of neuronal functions. Dehydroepiandrosterone (DHEA) exerts a neuroprotective effect by increasing the GABAergic tone through GABAA receptor stimulation. Therefore, we investigated the protective effect of DHEA in an animal model of HE, and the possible modulation of PKCε expression in different brain area.Methods: Fulminant hepatic failure was induced in 18 male, Sprague–Dawley rats by i.p. administration of 3 g/kg D-galactosamine, and after 30 min, a group of animals received a subcutaneous injection of 25 mg/kg (DHEA) repeated twice a day (3 days). Exploratory behavior and general activity were evaluated 24 h and 48 h after the treatments by the open field test. Then, brain cortex and cerebellum were used for immunoblotting analysis of PKCε level.Results: DHEA administration showed a significant improvement of locomotor activity both 24 and 48 h after D-galactosamine treatment (****p < 0.0001) but did not ameliorate liver parenchymal degeneration. Western blot analysis revealed a reduced immunoreactivity of PKCε (*p < 0.05) following D-galactosamine treatment in rat cortex and cerebellum. After the addition of DHEA, PKCε increased in the cortex in comparison with the D-galactosamine-treated (***p < 0.001) and control group (*p < 0.05), but decreased in the cerebellum (*p < 0.05) with respect to the control group. PKCε decreased after treatment with NH4Cl alone and in combination with DHEA in both cerebellum and cortex (****p < 0.0001). MTS assay demonstrated the synergistic neurotoxic action of NH4Cl and glutamate pretreatment in cerebellum and cortex along with an increased cell survival after DHEA pretreatment, which was significant only in the cerebellum (*p < 0.05).Conclusion: An association between the DHEA-mediated increase of PKCε expression and the improvement of comatose symptoms was observed. PKCε activation and expression in the brain could inhibit GABA-ergic tone counteracting HE symptoms. In addition, DHEA seemed to ameliorate the symptoms of HE and to increase the expression of PKCε in cortex and cerebellum.
- Published
- 2021
- Full Text
- View/download PDF
30. Iguratimod represses B cell terminal differentiation linked with the inhibition of PKC/EGR1 axis
- Author
-
Yan Ye, Mei Liu, Longhai Tang, Fang Du, Yuanhua Liu, Pei Hao, Qiong Fu, Qiang Guo, Qingran Yan, Xiaoming Zhang, and Chunde Bao
- Subjects
Iguratimod ,Rheumatoid arthritis (RA) ,Protein kinase C (PKC) ,Early growth response 1 (EGR1) ,Antibody-secreting cell (ASC) ,Diseases of the musculoskeletal system ,RC925-935 - Abstract
Abstract Background This study aimed to explore the molecular mechanism and clinical relevance of iguratimod in the regulation of human B cell terminal differentiation. Methods An in vitro human antibody-secreting cell (ASC) differentiation system was established to test the effect of iguratimod. B cell phenotype and key transcription factors (TFs) relevant to ASC differentiation were analyzed through flow cytometry and qPCR. The COX-2 activity was measured by enzyme immunoassay (EIA). RNA sequencing was used to identify potential targets of iguratimod. We enrolled six treatment-naive rheumatoid arthritis (RA) patients whose blood samples were collected for phenotypic and molecular studies along with 12-week iguratimod monotherapy. Results Iguratimod inhibited human ASC generation without affecting B cell activation and proliferation. Iguratimod showed only weak COX-2 activity. Gene set enrichment analysis (GSEA) identified that protein kinase C (PKC) pathway was targeted by iguratimod which was confirmed by PKC activity detection. Furthermore, early growth response 1 (EGR1), a target of PKC and a non-redundant TF for ASC differentiation, was found to be the most downregulated gene in iguratimod-treated B cells. Lastly, iguratimod monotherapy decreased peripheral ASCs and was associated with improved disease activity. The expression of major ASC-related TFs, including EGR1, was similarly downregulated in patient blood samples. Conclusions Iguratimod inhibits ASC differentiation both in vitro and in RA patients. Our study suggests that PKC/EGR1 axis, rather than COX-2, is critically involved in the inhibitory effect by iguratimod on human ASC differentiation. Iguratimod could have a broader application to treat B cell-related autoimmune diseases in clinics.
- Published
- 2019
- Full Text
- View/download PDF
31. Non-Alcoholic Fatty Liver Disease
- Author
-
Engin, Atilla, COHEN, IRUN R., Series editor, LAJTHA, ABEL, Series editor, LAMBRIS, JOHN D., Series editor, PAOLETTI, RODOLFO, Series editor, Engin, Ayse Basak, editor, and Engin, Atilla, editor
- Published
- 2017
- Full Text
- View/download PDF
32. Endothelial Dysfunction in Obesity
- Author
-
Engin, Atilla, COHEN, IRUN R., Series editor, LAJTHA, ABEL, Series editor, LAMBRIS, JOHN D., Series editor, PAOLETTI, RODOLFO, Series editor, Engin, Ayse Basak, editor, and Engin, Atilla, editor
- Published
- 2017
- Full Text
- View/download PDF
33. Mechanism of CNP-mediated DG-PKC and IP4 signaling pathway in diabetic rats with gastric motility disorder.
- Author
-
Lian, Hui-Ming, Guo, Jun-Yu, Sun, Yan, Zhang, Mo-Han, Piao, Li-Hua, Jin, Zheng, and Cai, Ying-Lan
- Abstract
In the precedent research conducted by the same team, it concluded that the activities in C-type natriuretic peptide (CNP)/cyclic guanosine monophosphate (cGMP)/cyclic adenosine monophosphate (cAMP)/β-type phospholipase C (PLCβ) pathways of rat antral smooth muscle were changed due to diabetes, which was the key pathogenetic mechanism for diabetic gastric dysmotility. As the follow-on step, this study was designed to probe into the downstream signaling pathway of CNP/PLCβ. The results showed that level of α-type protein kinase C (PKCα),cell membrane to cytoplasm ratio of PKCα, cell membrane to cytoplasmic ratio of βI-type protein kinase C (PKCβI) and level of Phosphor-PKCα (P-PKCα) were significantly reduced in diabetes rat antral smooth muscle samples. The content of tetraphosphate inositol (IP4) in gastric antral smooth muscle of diabetic rats reduced, and the content of diacyl-glycerol (DG) was unchanged. CNP significantly decreased the content of IP4 and DG, this effect was more obvious in diabetic rats. Subsequent to the addition of protein kinase A (PKA) blocker N-[2- (p-Bromocin-namylamino)ethyl]-5 -isoquinolinesulfonamide dihydrochloride (H-89) before CNP treatment, the inhibitory effect of CNP was reduced; subsequent to the addition of protein kinase G (PKG) blocker KT5823 before CNP treatment, the inhibitory effect of CNP was also reduced. With the addition of the combination of H-89 and KT5823 before CNP treatment, the inhibition by CNP could be offset. These results were concluded that CNP inhibited the activity of PKC family in rat smooth muscle and reduced the levels of IP4 and DG through the PKG/PKA-PLCβ pathways, causing inhibited muscular contractions, which may be a key pathogenetic factor for diabetic gastroparesis. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
34. Ingenane Diterpenoids
- Author
-
Appendino, Giovanni, Kinghorn, A. Douglas, Series editor, Falk, Heinz, Series editor, Gibbons, Simon, Series editor, and Kobayashi, Jun'ichi, Series editor
- Published
- 2016
- Full Text
- View/download PDF
35. Protein Kinase C Gene Fusions and Other Mechanisms for Loss of PKC Function in Cancer
- Author
-
Van, An-Angela Ngoc
- Subjects
Biochemistry ,Pharmacology ,Cellular biology ,autoinhibition ,cancer ,dominant-negative ,gene fusions ,loss-of-function ,protein kinase C (PKC) - Abstract
Protein kinase C (PKC) plays a critical role in cell signaling and homeostasis, regulating biological processes such as proliferation, differentiation, and apoptosis. Its dysregulation is associated with a multitude of pathophysiological states, with recent analyses of disease-associated mutations indicating that loss of PKC function is generally associated with cancer and gain of function with degenerative diseases. This dissertation expands on the mechanisms of PKC dysregulation in cancer, focusing on how gene fusions or mutations that disrupt autoinhibition cause loss of PKC. In the first part of the dissertation, newly identified PKC gene fusions in cancer encoding proteins that retain either the PKC catalytic or regulatory domain were characterized. Overexpression of catalytic domain fusions in cells revealed that they are constitutively active, as assessed using biosensors and other cellular assays. However, their inability to adopt an autoinhibited conformation resulted in their marked stability compared to full-length PKC. To assess whether these fusions were too unstable to accumulate at endogenous levels, CRISPR/Cas9-mediated gene editing was used to engineer a fusion of TANC2 with PRKCA. While the fusion mRNA was detected in the engineered cells, no detectable protein was expressed. Thus, the catalytic domain fusions are paradoxically loss-of-function. Characterization of a regulatory domain fusion revealed a dominant-negative role for the protein, suppressing the activity of wild-type PKC. The second part of the dissertation focused on additional mechanisms by which PKC is dysregulated in cancer. This work showed that [1] PKC that cannot be autoinhibited is subject to dephosphorylation by the phosphatase PHLPP and unphosphorylated PKC is sensitive to degradation; [2] inactivating mutants of PKC can be dominant-negative by sequestering common titratable components; and [3] impairment of the regulatory domains causes mislocalization of PKC. Taken together, these studies illustrate the diverse ways in which PKC function is lost in cancer, allowing cancer cells to overcome this cellular brake to tumor growth.
- Published
- 2020
36. Evidence for Angiotensin II as a Naturally Existing Suppressor for the Guanylyl Cyclase A Receptor and Cyclic GMP Generation
- Author
-
Xiao Ma, Seethalakshmi R. Iyer, Xiaoyu Ma, Shawn H. Reginauld, Yang Chen, Shuchong Pan, Ye Zheng, Dante G. Moroni, Yue Yu, Lianwen Zhang, Valentina Cannone, Horng H. Chen, Carlos M. Ferrario, S. Jeson Sangaralingham, and John C. Burnett
- Subjects
Inorganic Chemistry ,Organic Chemistry ,General Medicine ,Physical and Theoretical Chemistry ,Molecular Biology ,Spectroscopy ,Catalysis ,Computer Science Applications ,natriuretic peptide system (NPS) ,renin angiotensin aldosterone system (RAAS) ,angiotensin II (ANGII) ,cyclic guanosine monophosphate (cGMP) ,particulate guanylyl cyclase A (GC-A) ,ANGII type 1 receptor (AT1) ,protein kinase C (PKC) - Abstract
The natriuretic peptide system (NPS) and renin-angiotensin-aldosterone system (RAAS) function oppositely at multiple levels. While it has long been suspected that angiotensin II (ANGII) may directly suppress NPS activity, no clear evidence to date supports this notion. This study was designed to systematically investigate ANGII–NPS interaction in humans, in vivo, and in vitro. Circulating atrial, b-type, and c-type natriuretic peptides (ANP, BNP, CNP), cyclic guanosine monophosphate (cGMP), and ANGII were simultaneously investigated in 128 human subjects. Prompted hypothesis was validated in vivo to determine the influence of ANGII on ANP actions. The underlying mechanisms were further explored via in vitro approaches. In humans, ANGII demonstrated an inverse relationship with ANP, BNP, and cGMP. In regression models predicting cGMP, adding ANGII levels and the interaction term between ANGII and natriuretic peptides increased the predictive accuracy of the base models constructed with either ANP or BNP, but not CNP. Importantly, stratified correlation analysis further revealed a positive association between cGMP and ANP or BNP only in subjects with low, but not high, ANGII levels. In rats, co-infusion of ANGII even at a physiological dose attenuated cGMP generation mediated by ANP infusion. In vitro, we found the suppressive effect of ANGII on ANP-stimulated cGMP requires the presence of ANGII type-1 (AT1) receptor and mechanistically involves protein kinase C (PKC), as this suppression can be substantially rescued by either valsartan (AT1 blocker) or Go6983 (PKC inhibitor). Using surface plasmon resonance (SPR), we showed ANGII has low binding affinity to the guanylyl cyclase A (GC-A) receptor compared to ANP or BNP. Our study reveals ANGII is a natural suppressor for the cGMP-generating action of GC-A via AT1/PKC dependent manner and highlights the importance of dual-targeting RAAS and NPS in maximizing beneficial properties of natriuretic peptides in cardiovascular protection.
- Published
- 2023
- Full Text
- View/download PDF
37. 6,6′-Dihydroxythiobinupharidine (DTBN) Purified from Nuphar lutea Leaves Is an Inhibitor of Protein Kinase C Catalytic Activity
- Author
-
Kamran Waidha, Nikhil Ponnoor Anto, Divya Ram Jayaram, Avi Golan-Goldhirsh, Saravanakumar Rajendran, Etta Livneh, and Jacob Gopas
- Subjects
protein kinase C (PKC) ,Nuphar lutea ,6,6′-dihydroxythiobinupharidine (DTBN) ,kinase inhibitor ,Ramachandran plot ,homology docking modeling ,Organic chemistry ,QD241-441 - Abstract
Water lily (Nuphar) bioactive extracts have been widely used in traditional medicine owing to their multiple applications against human ailments. Phyto-active Nuphar extracts and their purified and synthetic derivatives have attracted the attention of ethnobotanists and biochemists. Here, we report that 6,6′-dihydroxythiobinupharidine (DTBN), purified from extracts of Nuphar lutea (L.) Sm. leaves, is an effective inhibitor of the kinase activity of members of the protein kinase C (PKC) family using in vitro and in silico approaches. We demonstrate that members of the conventional subfamily of PKCs, PKCα and PKCγ, were more sensitive to DTBN inhibition as compared to novel or atypical PKCs. Molecular docking analysis demonstrated the interaction of DTBN, with the kinase domain of PKCs depicting the best affinity towards conventional PKCs, in accordance with our in vitro kinase activity data. The current study reveals novel targets for DTBN activity, functioning as an inhibitor for PKCs kinase activity. Thus, this and other data indicate that DTBN modulates key cellular signal transduction pathways relevant to disease biology, including cancer.
- Published
- 2021
- Full Text
- View/download PDF
38. Signalling of Apoptin
- Author
-
Bullenkamp, Jessica, Tavassoli, Mahvash, Cohen, Irun R., Series editor, Lajtha, N.S. Abel, Series editor, Lambris, John D., Series editor, Paoletti, Rodolfo, Series editor, and Grimm, Stefan, editor
- Published
- 2014
- Full Text
- View/download PDF
39. eIF4G
- Author
-
Wagner, Simon D., Willis, Anne E., Beck, Daniel, and Parsyan, Armen, editor
- Published
- 2014
- Full Text
- View/download PDF
40. PKCγ and PKCε are Differentially Activated and Modulate Neurotoxic Signaling Pathways During Oxygen Glucose Deprivation in Rat Cortical Slices.
- Author
-
Surendran, Dayana
- Subjects
- *
PROTEIN kinase C , *CEREBRAL ischemia , *PROTEIN kinases , *INTRACELLULAR calcium , *SERINE/THREONINE kinases , *ISOENZYMES - Abstract
Cerebral ischemia is known to trigger a series of intracellular events such as changes in metabolism, membrane function and intracellular transduction, which eventually leads to cell death. Many of these processes are mediated by intracellular signaling cascades that involve protein kinase activation. Among all the kinases activated, the serine/threonine kinase family, protein kinase C (PKC), particularly, has been implicated in mediating cellular response to cerebral ischemic and reperfusion injury. In this study, using oxygen–glucose deprivation (OGD) in acute cortical slices as an in vitro model of cerebral ischemia, I show that PKC family of isozymes, specifically PKCγ and PKCε are differentially activated during OGD. Detecting the expression and activation levels of these isozymes in response to different durations of OGD insult revealed an early activation of PKCε and delayed activation of PKCγ, signifying their roles in response to different durations and stages of ischemic stress. Specific inhibition of PKCγ and PKCε significantly attenuated OGD induced cytotoxicity, rise in intracellular calcium, membrane depolarization and reactive oxygen species formation, thereby enhancing neuronal viability. This study clearly suggests that PKC family of isozymes; specifically PKCγ and PKCε are involved in OGD induced intracellular responses which lead to neuronal death. Thus isozyme specific modulation of PKC activity may serve as a promising therapeutic route for the treatment of acute cerebral ischemic injury. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
41. Expression and localization of grass carp pkc-θ (protein kinase C theta) gene after its activation.
- Author
-
Mehjabin, Rumana, Chen, Liangming, Huang, Rong, Zhu, Denghui, Yang, Cheng, Li, Yongming, Liao, Lanjie, He, Libo, Zhu, Zuoyan, and Wang, Yaping
- Subjects
- *
SERINE/THREONINE kinases , *PROTEIN kinase C , *CTENOPHARYNGODON idella - Abstract
Abstract Haemorrhagic disease caused by grass carp reovirus (GCRV) can result in large-scale death of young grass carp, leading to irreparable economic losses that seriously affect large-scale breeding. Protein kinase C (PKC, also known as PRKC) represents a family of serine/threonine protein kinases that includes multiple isozymes in many species. Among these, PKC-θ (PKC theta, also written as PRKCQ) is a novel isoform, mainly expressed in T cells, that is known to be involved in immune system function in mammals. To date, no research on immunological functions of fish Pkc-θ has been reported. To address this issue, we cloned the grass carp pkc-θ gene. Phylogenetic and syntenic analysis showed that this gene is the most evolutionarily conserved relative to zebrafish. Real-time quantitative PCR (RT-qPCR) indicated that pkc-θ was expressed at high levels in the gills and spleen of healthy grass carp. Infection with GCRV down regulated pkc-θ expression in the gills and spleen. Gene products that function upstream and downstream of pkc-θ were up regulated in the gill, but were down-regulated in the spleen. These results suggest that direct or indirect targeting of pkc-θ by GCRV may help the virus evade host immune defences in the spleen. Phorbol ester (PMA) treatment of Jurkat T cells induced translocation of grass carp Pkc-θ from the cytoplasm to the plasma membrane. This response to PMA suggests evolutionary conservation of an immune response function in fish Pkc-θ, as well as conservation of its sequence and structural domains. This study expanded our knowledge of the fish PKC gene family, and explored the role of pkc-θ in function of the grass carp immune system, providing new insights which may facilitate further studies of its biological functions. Highlights • Grass carp Pkc-θ was structurally conserved compared with other species. • Grass carp pkc-θ was enriched in the gill and spleen. • Grass carp pkc-θ and its related genes underwent similarily significant changes in gill and spleen after GCRV infection. • Phorbol ester (PMA) made grass carp Pkc-θ translocated from- cytoplasm to the membrane in Jurkat T cells. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
42. PKCα-Mediated Downregulation of RhoA Activity in Depolarized Vascular Smooth Muscle: Synergistic Vasorelaxant Effect of PKCα and ROCK Inhibition.
- Author
-
del Carmen González-Montelongo, María, Porras-González, Cristina, González-Montelongo, Rafaela, Revilla-González, Gonzalo, Dolores Pastor, María, Castellano, Antonio, and Ureña, Juan
- Subjects
- *
DOWNREGULATION , *VASCULAR smooth muscle , *PROTEIN kinase C , *MASS spectrometry , *SMALL interfering RNA , *CELL membranes - Abstract
Background/Aims: Protein kinase C (PKC)- and RhoA/Rho-associated kinase (ROCK) play important roles in arterial sustained contraction. Although depolarization-elicited RhoA/ROCK activation is accepted, the role of PKC in depolarized vascular smooth muscle cells (VSMCs) is a subject of controversy. Our aim was to study the role of PKC in arterial contraction and its interaction with RhoA/ROCK. Methods: Mass spectrometry was used to identify the PKC isoenzymes. PKCα levels and RhoA activity were analyzed by western blot and G-LISA, respectively, and isometric force was measured in arterial rings. Results: In depolarized VSMCs RhoA and PKCα were translocated to the plasma membrane, where they colocalize and coimmunoprecipitate. Interestingly, depolarization-induced RhoA activation was downregulated by PKCα, effect reverted by PKCα inhibition. Phorbol 12,13-dibutyrate (PDBu) induced the translocation of PKCα to the plasma membrane, increased the level of RhoA in the cytosol and reduced RhoA/ROCK activity. These effects were reverted when PKC was inhibited. Pharmacological or siRNA inhibition of PKCα synergistically potentiated the vasorelaxant effect of RhoA/ROCK inhibition. Conclusion: The present study provides the first evidence that RhoA activity is downregulated by PKCα in depolarized and PDBu treated freshly isolated VSMCs and arteries, with an important physiological role on arterial contractility. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
43. Extracellular Histone-Induced Protein Kinase C Alpha Activation and Troponin Phosphorylation Is a Potential Mechanism of Cardiac Contractility Depression in Sepsis
- Author
-
Simon T. Abrams, Yasir Alhamdi, Min Zi, Fengmei Guo, Min Du, Guozheng Wang, Elizabeth J. Cartwright, and Cheng-Hock Toh
- Subjects
Inorganic Chemistry ,sepsis ,troponin ,phosphorylation ,cardiac contractility ,protein kinase C (PKC) ,Organic Chemistry ,General Medicine ,Physical and Theoretical Chemistry ,Molecular Biology ,Spectroscopy ,Catalysis ,Computer Science Applications - Abstract
Reduction in cardiac contractility is common in severe sepsis. However, the pathological mechanism is still not fully understood. Recently it has been found that circulating histones released after extensive immune cell death play important roles in multiple organ injury and disfunction, particularly in cardiomyocyte injury and contractility reduction. How extracellular histones cause cardiac contractility depression is still not fully clear. In this work, using cultured cardiomyocytes and a histone infusion mouse model, we demonstrate that clinically relevant histone concentrations cause significant increases in intracellular calcium concentrations with subsequent activation and enriched localization of calcium-dependent protein kinase C (PKC) α and βII into the myofilament fraction of cardiomyocytes in vitro and in vivo. Furthermore, histones induced dose-dependent phosphorylation of cardiac troponin I (cTnI) at the PKC-regulated phosphorylation residues (S43 and T144) in cultured cardiomyocytes, which was also confirmed in murine cardiomyocytes following intravenous histone injection. Specific inhibitors against PKCα and PKCβII revealed that histone-induced cTnI phosphorylation was mainly mediated by PKCα activation, but not PKCβII. Blocking PKCα also significantly abrogated histone-induced deterioration in peak shortening, duration and the velocity of shortening, and re-lengthening of cardiomyocyte contractility. These in vitro and in vivo findings collectively indicate a potential mechanism of histone-induced cardiomyocyte dysfunction driven by PKCα activation with subsequent enhanced phosphorylation of cTnI. These findings also indicate a potential mechanism of clinical cardiac dysfunction in sepsis and other critical illnesses with high levels of circulating histones, which holds the potential translational benefit to these patients by targeting circulating histones and downstream pathways.
- Published
- 2023
- Full Text
- View/download PDF
44. Receptor Signaling Integration by TRP Channelsomes
- Author
-
Mori, Yasuo, Kajimoto, Taketoshi, Nakao, Akito, Takahashi, Nobuaki, Kiyonaka, Shigeki, and Islam, Md. Shahidul, editor
- Published
- 2011
- Full Text
- View/download PDF
45. Removal of Potential Phosphorylation Sites does not Alter Creatine Transporter Response to PKC or Substrate Availability
- Author
-
Lucia Santacruz, Marcus D. Darrabie, Rajashree Mishra, and Danny O. Jacobs
- Subjects
Energy metabolism ,Cardiac metabolism ,Creatine ,Membrane transport ,Phosphorylation ,Protein kinase C (PKC) ,Physiology ,QP1-981 ,Biochemistry ,QD415-436 - Abstract
Background: Creatine, Phosphocreatine, and creatine kinases, constitute an energy shuttle that links ATP production in mitochondria with cellular consumption sites. Myocytes and neurons cannot synthesize creatine and depend on uptake across the cell membrane by a specialized transporter to maintain intracellular creatine levels. Although recent studies have improved our understanding of creatine transport in cardiomyocytes, the structural elements underlying the creatine transporter protein regulation and the relevant intracellular signaling processes are unknown. Methods: The effects of pharmacological activation of kinases or phosphatases on creatine transport in cardiomyocytes in culture were evaluated. Putative phosphorylation sites in the creatine transporter protein were identified by bioinformatics analyses, and ablated using site-directed mutagenesis. Mutant transporter function and their responses to pharmacological PKC activation or changes in creatine availability in the extracellular environment, were evaluated. Results: PKC activation decreases creatine transport in cardiomyocytes in culture. Elimination of high probability potential phosphorylation sites did not abrogate responses to PKC activation or substrate availability. Conclusion: Modulation of creatine transport in cardiomyocytes is a complex process where phosphorylation at predicted sites in the creatine transporter protein does not significantly alter activity. Instead, non-classical structural elements in the creatine transporter and/or interactions with regulatory subunits may modulate its activity.
- Published
- 2015
- Full Text
- View/download PDF
46. Signal Cross Talks for Sustained MAPK Activation and Cell Migration Mediated by Reactive Oxygen Species: The Involvement in Tumor Progression
- Author
-
Hu, Chi-Tan, Wu, Jia-Ru, Wu, Wen-Sheng, Wu, Wen-Sheng, editor, and Hu, Chi-Tan, editor
- Published
- 2010
- Full Text
- View/download PDF
47. Different roles of cAMP/PKA and PKC signaling in regulating progesterone and PGE2 levels in immortalized rat granulosa cell cultures.
- Author
-
Nemer, Ala, Azab, Abed N., Rimon, Gilad, Lamprecht, Sergio, and Ben-Menahem, David
- Subjects
- *
PROGESTERONE , *GRANULOSA cell tumors , *FOLLICULAR dendritic cells , *ANTIGEN presenting cells , *CYCLIC-AMP-dependent protein kinase - Abstract
Highlights • Immortalized rat granulosa cells isolated from pre ovulatory follicles produce PGE 2. • PMSG, forskolin and a cAMP analog increase progesterone but not PGE 2 media levels. • In contrast, TPA stimulates PGE 2 but not progesterone secretion from the cells. • PKA and PKC have opposite effects on progesterone and PGE 2 secretion from the cells. • This may be beneficial to coordinate two key mediators in the preovulatory follicle. Abstract Follicular cells from various species secrete steroids and prostaglandins, which are crucial for reproduction, in response to gonadotropins. Here, we examined prostaglandin E 2 (PGE 2) secretion from immortalized rat granulosa cells derived from preovulaotry follicles expressing the rat follicle stimulating hormone receptor (denoted as FSHR cells) that produce progesterone in response to gonadotropins. The cells were stimulated with a) pregnant mare's serum gonadotropin (PMSG; a rat FSH receptor agonist), b) activators of the protein kinase A (PKA) pathway (forskolin and a cell permeable cAMP analog Dibutyryl-cAMP (DB-cAMP)) and c) protein kinase C (PKC) (12-O-tetradecanoylphorbol 13-acetate; TPA), alone and in combination for 24 h. Thereafter, PGE 2 and progesterone levels in the culture media were determined. In accordance with previous studies, while PMSG and the PKA pathway activators induced progesterone accumulation in the media, TPA did not. In contrast, our data indicate that TPA, but neither PMSG, forskolin and DB-cAMP evoked PGE 2 accumulation in the media. Western Blot analysis of cell lysate showed a drastic TPA induced increase of COX-2 levels, which was not seen with neither PMSG nor forskolin treatment. This association between the COX-2 and PGE 2 levels suggests that the enzyme activity is the likely factor that determines the synthesis and levels of the prostaglandin in the culture media of the granulosa-derived cells. The addition of the PKA inhibitor H-89 to the FSHR cultures suppressed the gonadotropin and forskolin induction of progesterone secretion. Incubation in the presence of GF109203X (a PKC inhibitor) attenuated the TPA induced PGE 2 accumulation in the culture media of the cells (a dose dependent reduction of 40–70%). In addition, while TPA inhibited the PMSG and forskolin induced-accumulation of progesterone in the media, the gonadotropin and forskolin inhibited the elevation of PGE 2 levels evoked by TPA (a dose dependent decrease of 35–55%). These data suggest that cAMP/PKA and PKC signaling have opposite effects on PGE 2 and progesterone synthesis in FSHR cells. We propose that this PKA and PKC interplay on progesterone and PGE 2 may be advantageous for the coordination of these key mediators for successful ovulation and luteinization. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
48. Suppressing PKC-dependent membrane P2X3 receptor upregulation in dorsal root ganglia mediated electroacupuncture analgesia in rat painful diabetic neuropathy.
- Author
-
Zhou, Ya-feng, Ying, Xiao-ming, He, Xiao-fen, Shou, Sheng-Yun, Wei, Jun-Jun, Tai, Zhao-xia, Shao, Xiao-mei, Liang, Yi, Fang, Fang, Fang, Jian-qiao, and Jiang, Yong-liang
- Abstract
Painful diabetic neuropathy (PDN) is a common and troublesome diabetes complication. Protein kinase C (PKC)-mediated dorsal root ganglia (DRG) P2X3 receptor upregulation is one important mechanism underlying PDN. Accumulating evidence demonstrated that electroacupuncture (EA) at low frequency could effectively attenuate neuropathic pain. Our previous study showed that 2-Hz EA could relieve pain well in PDN. The study aimed to investigate whether 2-Hz EA relieves pain in PDN through suppressing PKC-mediated DRG P2X3 receptor upregulation. A 7-week feeding of high-fat and high-sugar diet plus a single injection of streptozotocin (STZ) in a dose of 35 mg/kg after a 5-week feeding of the diet successfully induced type 2 PDN in rats as revealed by the elevated body weight, fasting blood glucose, fasting insulin and insulin resistance, and the reduced paw withdrawal threshold (PWT), as well as the destructive ultrastructural change of sciatic nerve. DRG plasma membrane P2X3 receptor level and DRG PKC expression were elevated. Two-hertz EA failed to improve peripheral neuropathy; however, it reduced PWT, DRG plasma membrane P2X3 receptor level, and DRG PKC expression in PDN rats. Intraperitoneal administration of P2X3 receptor agonist αβ-meATP or PKC activator phorbol 12-myristate 13-acetate (PMA) blocked 2-Hz EA analgesia. Furthermore, PMA administration increased DRG plasma membrane P2X3 receptor level in PDN rats subject to 2-Hz EA treatment. These findings together indicated that the analgesic effect of EA in PDN is mediated by suppressing PKC-dependent membrane P2X3 upregulation in DRG. EA at low frequency is a valuable approach for PDN control. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
49. Protein kinase C mediated internalization of ErbB2 is independent of clathrin, ubiquitination and Hsp90 dissociation.
- Author
-
Malik, Muhammad Salman, Nikolaysen, Filip, Stang, Espen, Dietrich, Markus, and Skeie, Marianne
- Subjects
- *
EPIDERMAL growth factor receptors , *GENETIC overexpression , *PROTEIN kinase C genetics , *CLATHRIN , *UBIQUITINATION , *HEAT shock proteins - Abstract
Abstract Overexpression of ErbB2 is frequent in cancer and understanding the mechanisms which regulate its expression is important. ErbB2 is considered endocytosis resistant. It has no identified ligand, but upon heterodimerization it is a potent mediator of proliferative signaling. A recent study established a role for protein kinase C (PKC) in internalization and recycling of ErbB2. We have now further investigated the molecular mechanisms involved in PKC-mediated downregulation of ErbB2. We confirm that PMA-induced PKC activation causes ErbB2 internalization, but while the Hsp90 inhibitor 17-AAG induced ErbB2 degradation, PMA had no such effect. When combined with 17-AAG, PMA had additive effect on ErbB2 internalization indicating that Hsp90 inhibition and PKC activation induce internalization by alternative mechanisms. We confirm that while 17-AAG-induced internalization was clathrin-mediated, PMA-induced internalization was clathrin independent. This difference may be explained by while both 17-AAG and PMA reduced the constitutive tyrosine phosphorylation of ErbB2, only 17-AAG induced Hsp90 dissociation, Hsp70 recruitment and ubiquitination of ErbB2. Importantly, since PMA induced internalization of ErbB2, but not dissociation of Hsp90, Hsp90 does not per se retain ErbB2 at the plasma membrane. The morphology of the compartment into which receptors are sorted upon PKC activation has not previously been identified. By immuno-electron microscopy, we show that PMA sorts ErbB2 into a complex tubulovesicular or cisternal organelle resembling a previously described endocytic recycling compartment. Highlights • PKC activation induces internalization but not degradation of ErbB2. • PKC-induced internalization of ErbB2 is independent of Hsp90 dissociation. • PKC-induced internalization of ErbB2 is independent of ErbB2 ubiquitination. • PKC-induced internalization of ErbB2 is clathrin independent. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
50. Exogenous CGRP upregulates profibrogenic growth factors through PKC/JNK signaling pathway in kidney proximal tubular cells.
- Author
-
Yoon, Sang Pil and Kim, Jinu
- Abstract
Kidney denervation prevents the development of tubulointerstitial fibrosis, but the neuropeptide calcitonin gene-related peptide (CGRP) in the denervated kidneys restores the fibrotic feature through the upregulation of profibrogenic growth factors. CGRP is involved in aggravation of inflammation by increasing the number of circulating cells and chemotactic factors. However, it is not clear how CGRP contributes to the upregulation of profibrogenic factors during fibrogenesis. In both human and pig kidney proximal tubular cell lines, administration of 1 nM CGRP significantly increased the levels of transforming growth factor-β1 (TGF-β1) production and connective tissue growth factor (CTGF) expression at 6 and 24 h after the administration. Exogenous CGRP also increased the TGF-β1 and CTGF protein levels in the incubation media, indicating release of these proteins from the cells. Treatment with 100 nM CGRP receptor antagonist (CGRP
8-37 ) for 24 h significantly inhibited the increase in intracellular levels and released levels of TGF-β1 and CTGF in CGRP-treated cells. Genetic inhibition of CGRP receptor using siRNA transfection also suppressed the increase in TGF-β1 production and release at 24 h after CGRP stimulation. Furthermore, treatment with a specific protein kinase C (PKC) inhibitor chelerythrine (1 thru 10 μM) markedly reduced the upregulation and release of TGF-β1 and CTGF 6 h after CGRP administration. Finally, inhibition of c-Jun N-terminal protein kinase (JNK) phosphorylation using 1 μM SP600125 prevented the increase in TGF-β1 and CTGF upregulation and release 6 h after CGRP administration. Consistent with the in vitro data, exogenous CGRP in denervated UUO kidneys upregulated and secreted TGF-β1 and CTGF in dependence on PKC activation and JNK phosphorylation. In conclusion, these data suggest that exogenous CGRP induces the upregulation and secretion of profibrogenic TGF-β1 and CTGF proteins through the CGRP receptor/PKC/JNK signaling pathway in kidney proximal tubular cells. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.