1
|
Xie Y, Liang B, Meng Z, Guo R, Liu C, Yuan Y, Mu W, Wang Y, Cao J. Downregulation of HSPB1 and MGST1 Promotes Ferroptosis and Impacts Immune Infiltration in Diabetic Cardiomyopathy. Cardiovasc Toxicol 2025; 25:719-734. [PMID: 40053272 DOI: 10.1007/s12012-025-09982-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 02/26/2025] [Indexed: 03/12/2025]
Abstract
Diabetic cardiomyopathy (DCM) is a leading cause of death in diabetic patients. Current therapies do not adequately resolve this problem and focus only on the optimal level of blood glucose for patients. Ferroptosis plays an important role in diabetes mellitus and cardiovascular diseases. However, the role of ferroptosis in DCM remains unclear. Differentially expressed ferroptosis-related genes (DE-FRGs) were identified by intersection of the GSE26887 dataset and the Ferroptosis Database. The associations between the DE-FRGs and immune cells in DCM, estimated via the CIBERSORTx algorithm, were analysed. Flow cytometry (FCM) was used to evaluate the infiltration of immune cells in myocardial tissues. The expression of DE-FRGs, glutathione peroxidase 4 and solute carrier family 7 member 11 was examined via real-time quantitative PCR and Western blotting. Three DE-FRGs were identified: heat shock protein family B (small) member 1 (HSPB1), microsomal glutathione S-transferase 1 (MGST1) and solute carrier family 40 member 1 (SLC40A1), which are closely linked to immune cells in DCM. In vivo, the levels of CD8 + T cells, B cells and regulatory T (Treg) cells were significantly decreased in the DCM group, whereas the levels of CD4 + T cells, M1 cells, M2 cells and monocytes were increased. Diabetes significantly decreased HSPB1 and MGST1 levels and increased ferroptosis compared with the Normal group. Furthermore, the ferroptosis inhibitor ferrostatin-1 (Fer-1) alleviated high-fat diet (HFD)-induced cardiomyocyte injury and rescued ferroptosis. These findings suggest that the ferroptosis-related genes HSPB1 and MGST1 are closely related to immune cell infiltration and may be therapeutic targets for DCM.
Collapse
MESH Headings
- Ferroptosis/drug effects
- Animals
- Diabetic Cardiomyopathies/immunology
- Diabetic Cardiomyopathies/genetics
- Diabetic Cardiomyopathies/pathology
- Diabetic Cardiomyopathies/enzymology
- Diabetic Cardiomyopathies/metabolism
- Male
- Down-Regulation
- Myocytes, Cardiac/pathology
- Myocytes, Cardiac/immunology
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Disease Models, Animal
- Molecular Chaperones/genetics
- Molecular Chaperones/metabolism
- Humans
- Signal Transduction
- Diabetes Mellitus, Experimental/enzymology
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/immunology
- HSP27 Heat-Shock Proteins/genetics
- HSP27 Heat-Shock Proteins/metabolism
- Mice, Inbred C57BL
- Heat-Shock Proteins/genetics
- Heat-Shock Proteins/metabolism
- Databases, Genetic
Collapse
Affiliation(s)
- Yaoli Xie
- MOE Key Laboratory of Cellular Physiology and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Bin Liang
- MOE Key Laboratory of Cellular Physiology and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Zhijun Meng
- MOE Key Laboratory of Cellular Physiology and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Rui Guo
- MOE Key Laboratory of Cellular Physiology and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Caihong Liu
- MOE Key Laboratory of Cellular Physiology and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Yi Yuan
- MOE Key Laboratory of Cellular Physiology and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Wei Mu
- Department of Interventional Therapy for Tumor and Vascular Interventional Therapy, The Fifth Hospital of Shanxi Medical University (Shanxi Provincial People's Hospital), Taiyuan, China
| | - Yajing Wang
- Department of Biomedical Engineering, Heersink School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| | - Jimin Cao
- MOE Key Laboratory of Cellular Physiology and the Department of Physiology, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
2
|
Xie Y, Liang B, Meng Z, Guo R, Liu C, Yuan Y, Mu W, Wang Y, Cao J. Down-regulation of HSPB1 and MGST1 promote ferroptosis and impact immune infiltration in diabetic cardiomyopathy. RESEARCH SQUARE 2024:rs.3.rs-5153598. [PMID: 39711549 PMCID: PMC11661379 DOI: 10.21203/rs.3.rs-5153598/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Diabetic cardiomyopathy (DCM) is a leading cause of death in diabetic patients. Current therapies do not adequately resolve this problem and focus only on the optimal level of blood glucose for patients. Ferroptosis plays an important role in diabetes mellitus and cardiovascular diseases. However, the role of ferroptosis in DCM remains unclear. Differentially expressed ferroptosis-related genes (DE-FRGs) were identified by intersecting GSE26887 dataset and the Ferroptosis Database (FerrDb). The associations between the DE-FRGs and immune cells in DCM, estimated by CIBERSORTx algorithm, were analyzed. Using ow cytometry (FCM) to evaluated the infiltration of immune cells of myocardial tissues. The expression of DE-FRGs, Glutathione peroxidase 4 (GPX4) and Solute carrier family 7 member 11 (SLC7A11) were examined by real-time quantitative PCR and western blotting. 3 DE-FRGs were identified, which are Heat shock protein family B (small) member 1 (HSPB1), Microsomal glutathione S-transferase 1 (MGST1) and solute carrier family 40 member 1 (SLC40A1) respectively, and they were closely linked to immune cells in DCM. In vivo, the levels of CD8 + T cells, B cells and Treg cells were significantly decreased in the DCM group, while the levels of CD4 + T cells, M1 cells, M2 cells and monocytes were increased. Diabetes significantly decreased HSPB1 and MGST1 levels and increased ferroptosis compared to normal group. Furthermore, ferroptosis inhibitor ferrostatin-1 (Fer-1) alleviated high-fat diet (HFD)-induced cadiomyocyte injury and rescued the ferroptosis. This study suggests that ferroptosis related gene HSPB1 and MGST1 are closely related to immune cell infiltration, which may become therapeutic targets for DCM.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wei Mu
- The Fifth Hospital of Shanxi Medical University (Shanxi Provincial People's Hospital)
| | | | | |
Collapse
|
3
|
Yang C, Zhao X, An X, Zhang Y, Sun W, Zhang Y, Duan Y, Kang X, Sun Y, Jiang L, Lian F. Axonal transport deficits in the pathogenesis of diabetic peripheral neuropathy. Front Endocrinol (Lausanne) 2023; 14:1136796. [PMID: 37056668 PMCID: PMC10086245 DOI: 10.3389/fendo.2023.1136796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
Diabetic peripheral neuropathy (DPN) is a chronic and prevalent metabolic disease that gravely endangers human health and seriously affects the quality of life of hyperglycemic patients. More seriously, it can lead to amputation and neuropathic pain, imposing a severe financial burden on patients and the healthcare system. Even with strict glycemic control or pancreas transplantation, peripheral nerve damage is difficult to reverse. Most current treatment options for DPN can only treat the symptoms but not the underlying mechanism. Patients with long-term diabetes mellitus (DM) develop axonal transport dysfunction, which could be an important factor in causing or exacerbating DPN. This review explores the underlying mechanisms that may be related to axonal transport impairment and cytoskeletal changes caused by DM, and the relevance of the latter with the occurrence and progression of DPN, including nerve fiber loss, diminished nerve conduction velocity, and impaired nerve regeneration, and also predicts possible therapeutic strategies. Understanding the mechanisms of diabetic neuronal injury is essential to prevent the deterioration of DPN and to develop new therapeutic strategies. Timely and effective improvement of axonal transport impairment is particularly critical for the treatment of peripheral neuropathies.
Collapse
|
4
|
Hashimoto-Torii K, Sasaki M, Chang YW, Hwang H, Waxman SG, Kocsis JD, Rakic P, Torii M. Detection of local and remote cellular damage caused by spinal cord and peripheral nerve injury using a heat shock signaling reporter system. IBRO Rep 2018; 5:91-98. [PMID: 30480161 PMCID: PMC6240805 DOI: 10.1016/j.ibror.2018.11.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 11/05/2018] [Indexed: 12/14/2022] Open
Abstract
Spinal cord and peripheral nerve injury results in extensive damage to the locally injured cells as well as distant cells that are functionally connected to them. Both primary and secondary damage can cause a broad range of clinical abnormalities, including neuropathic pain and cognitive and memory dysfunction. However, the mechanisms underlying these abnormalities remain unclear, awaiting new methods to identify affected cells to enable examination of their molecular, cellular and physiological characteristics. Here, we report that both primary and secondary damage to cells in mouse models of spinal cord and peripheral nerve injury can be detected in vivo using a novel fluorescent reporter system based on the immediate stress response via activation of Heat Shock Factor 1. We also provide evidence for altered electrophysiological properties of reporter-positive secondarily-injured neurons. The comprehensive identification of injured, but surviving cells located both close and at distant locations from the injury site in vivo will provide a way to study their pathophysiology and possibly prevention of their further deterioration.
Collapse
Key Words
- Cellular damage
- DRG, dorsal root ganglion
- FG, Fluoro-Gold
- HRP, horseradish peroxidase
- HSE, heat shock-response element
- HSF1, heat shock factor 1
- HSP, heat shock protein
- Heat shock signaling
- IL-6, interleukin 6
- M1, primary motor cortex
- M2, secondary motor cortex
- MPtA, medial parietal association cortex
- PBS, phosphate buffered saline
- PCR, polymerase chain reaction
- RFP, red fluorescent protein
- Reporter mouse
- SCI, spinal cord injury
- SNI, sciatic nerve injury
- Sciatic nerve injury
- Spinal cord injury
- WDR, wide-dynamic range
- WGA, wheat germ agglutinin
Collapse
Affiliation(s)
- Kazue Hashimoto-Torii
- Center for Neuroscience Research, Children’s Research Institute, Children’s National Medical Center, Washington, DC 20010, USA
- Department of Pediatrics, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
- Department of Neurobiology and Kavli Institute for Neuroscience, School of Medicine, Yale University, New Haven, CT 06510, USA
| | - Masanori Sasaki
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, 06510, USA
- Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, Connecticut, 06516, USA
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Hokkaido, 060-8556, Japan
| | - Yu-Wen Chang
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, 06510, USA
- Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, Connecticut, 06516, USA
| | - Hye Hwang
- Center for Neuroscience Research, Children’s Research Institute, Children’s National Medical Center, Washington, DC 20010, USA
- Institute of Biomedical Sciences, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Stephen G. Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, 06510, USA
- Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, Connecticut, 06516, USA
| | - Jeffery D. Kocsis
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, 06510, USA
- Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, Connecticut, 06516, USA
| | - Pasko Rakic
- Department of Neurobiology and Kavli Institute for Neuroscience, School of Medicine, Yale University, New Haven, CT 06510, USA
| | - Masaaki Torii
- Center for Neuroscience Research, Children’s Research Institute, Children’s National Medical Center, Washington, DC 20010, USA
- Department of Pediatrics, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
- Department of Neurobiology and Kavli Institute for Neuroscience, School of Medicine, Yale University, New Haven, CT 06510, USA
| |
Collapse
|
5
|
Nafar F, Williams JB, Mearow KM. Astrocytes release HspB1 in response to amyloid-β exposure in vitro. J Alzheimers Dis 2016; 49:251-63. [PMID: 26444769 DOI: 10.3233/jad-150317] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Although heat shock proteins are thought to function primarily as intracellular chaperones, the release and potential extracellular functions of heat shock proteins have been the focus of an increasing number of studies. Our particular interest is HspB1 (Hsp25/27) and as astrocytes are an in vivo source of HspB1 it is a reasonable possibility they could release HspB1 in response to local stresses. Using primary cultures of rat cortical astrocytes, we investigated the extracellular release of HspB1 with exposure to amyloid-β (Aβ). In order to assess potential mechanisms of release, we cotreated the cells with compounds that can modulate protein secretion including Brefeldin A, Methyl β-cyclodextrin, and MAP kinase inhibitors. Exposure to Aβ (0.1, 1.0, 2.0 μM) for 24-48 h resulted in a selective release of HspB1 that was insensitive to BFA treatment; none of the other inhibitors had any detectable influence. Protease protection assays indicated that some of the released HspB1 was associated with a membrane bound fraction, and analysis of exosomal preparations indicated the presence of HspB1 in exosomes. Finally, immunoprecipitation experiments demonstrated that the extracellular HspB1 was able to interact with extracellular Aβ. In summary, Aβ can stimulate release of HspB1 from astrocytes, this release is insensitive to Golgi or lipid raft disruption, and HspB1 can be found either free in the medium or associated with exosomes. This release suggests that there is a potential for extracellular HspB1 to be able to bind and sequester extracellular Aβ.
Collapse
|
6
|
Apara A, Goldberg JL. Molecular mechanisms of the suppression of axon regeneration by KLF transcription factors. Neural Regen Res 2014; 9:1418-21. [PMID: 25317150 PMCID: PMC4192940 DOI: 10.4103/1673-5374.139454] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2014] [Indexed: 01/11/2023] Open
Abstract
Molecular mechanisms of the Krüppel-like family of transcription factors (KLFs) have been studied more in proliferating cells than in post-mitotic cells such as neurons. We recently found that KLFs regulate intrinsic axon growth ability in central nervous system (CNS) neurons including retinal ganglion cells, and hippocampal and cortical neurons. With at least 15 of 17 KLF family members expressed in neurons and at least 5 structurally unique subfamilies, it is important to determine how this complex family functions in neurons to regulate the intricate genetic programs of axon growth and regeneration. By characterizing the molecular mechanisms of the KLF family in the nervous system, including binding partners and gene targets, and comparing them to defined mechanisms defined outside the nervous system, we may better understand how KLFs regulate neurite growth and axon regeneration.
Collapse
Affiliation(s)
| | - Jeffrey L Goldberg
- Shiley Eye Center, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
7
|
Li L, Zuo Y, He J. Preconditioning crush increases the survival rate of motor neurons after spinal root avulsion. Neural Regen Res 2014. [PMID: 25206852 PMCID: PMC4153498 DOI: 10.4103/1673-5374.130096] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In a previous study, heat shock protein 27 was persistently upregulated in ventral motor neurons following nerve root avulsion or crush. Here, we examined whether the upregulation of heat shock protein 27 would increase the survival rate of motor neurons. Rats were divided into two groups: an avulsion-only group (avulsion of the L4 lumbar nerve root only) and a crush-avulsion group (the L4 lumbar nerve root was crushed 1 week prior to the avulsion). Immunofluorescent staining revealed that the survival rate of motor neurons was significantly greater in the crush-avulsion group than in the avulsion-only group, and this difference remained for at least 5 weeks after avulsion. The higher neuronal survival rate may be explained by the upregulation of heat shock protein 27 expression in motor neurons in the crush-avulsion group. Furthermore, preconditioning crush greatly attenuated the expression of nitric oxide synthase in the motor neurons. Our findings indicate that the neuroprotective action of preconditioning crush is mediated through the upregulation of heat shock protein 27 expression and the attenuation of neuronal nitric oxide synthase upregulation following avulsion.
Collapse
Affiliation(s)
- Lin Li
- Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yizhi Zuo
- Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jianwen He
- Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
8
|
Pourhamidi K, Skärstrand H, Dahlin LB, Rolandsson O. HSP27 concentrations are lower in patients with type 1 diabetes and correlate with large nerve fiber dysfunction. Diabetes Care 2014; 37:e49-50. [PMID: 24558083 DOI: 10.2337/dc13-1780] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
9
|
de Thonel A, Le Mouël A, Mezger V. Transcriptional regulation of small HSP-HSF1 and beyond. Int J Biochem Cell Biol 2012; 44:1593-612. [PMID: 22750029 DOI: 10.1016/j.biocel.2012.06.012] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Revised: 06/07/2012] [Accepted: 06/08/2012] [Indexed: 12/16/2022]
Abstract
The members of the small heat shock protein (sHSP) family are molecular chaperones that play major roles in development, stress responses, and diseases, and have been envisioned as targets for therapy, particularly in cancer. The molecular mechanisms that regulate their transcription, in normal, stress, or pathological conditions, are characterized by extreme complexity and subtlety. Although historically linked to the heat shock transcription factors (HSFs), the stress-induced or developmental expression of the diverse members, including HSPB1/Hsp27/Hsp25, αA-crystallin/HSPB4, and αB-crystallin/HSPB5, relies on the combinatory effects of many transcription factors. Coupled with remarkably different cis-element architectures in the sHsp regulatory regions, they confer to each member its developmental expression or stress-inducibility. For example, multiple regulatory pathways coordinate the spatio-temporal expression of mouse αA-, αB-crystallin, and Hsp25 genes during lens development, through the action of master genes, like the large Maf family proteins and Pax6, but also HSF4. The inducibility of Hsp27 and αB-crystallin transcription by various stresses is exerted by HSF-dependent mechanisms, by which concomitant induction of Hsp27 and αB-crystallin expression is observed. In contrast, HSF-independent pathways can lead to αB-crystallin expression, but not to Hsp27 induction. Not surprisingly, deregulation of the expression of sHSP is associated with various pathologies, including cancer, neurodegenerative, or cardiac diseases. However, many questions remain to be addressed, and further elucidation of the developmental mechanisms of sHsp gene transcription might help to unravel the tissue- and stage-specific functions of this fascinating class of proteins, which might prove to be crucial for future therapeutic strategies. This article is part of a Directed Issue entitled: Small HSPs in physiology and pathology.
Collapse
|
10
|
Pourhamidi K, Dahlin LB, Boman K, Rolandsson O. Heat shock protein 27 is associated with better nerve function and fewer signs of neuropathy. Diabetologia 2011; 54:3143-9. [PMID: 21909836 DOI: 10.1007/s00125-011-2303-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Accepted: 08/11/2011] [Indexed: 01/09/2023]
Abstract
AIMS/HYPOTHESIS High levels of serum heat shock protein 27 (sHSP27) have been associated with distal symmetric polyneuropathy in patients with type 1 diabetes. Our objective was to investigate the association between sHSP27, neuropathic signs and nerve function in individuals with normal glucose tolerance (NGT), impaired glucose tolerance (IGT) and type 2 diabetes. METHODS Participants were recruited consecutively from the population-based Västerbotten Intervention Program (NGT, n = 39, IGT, n = 29, and type 2 diabetes, n = 51) and were matched for age and sex. sHSP27 levels were measured and nerve conduction studies were performed (peroneal and sural nerves). z Scores for each nerve conduction measure were calculated and compiled into a composite z score for the leg. Neuropathy disability score (NDS) was used to assess neuropathic signs. RESULTS Patients with diabetes had significantly lower sHSP27 levels (geometric mean sHSP27 206 pg/ml, 95% CI 142, 299) than those with IGT (geometric mean sHSP27 455 pg/ml, 95% CI 319, 650, p < 0.05) and controls (geometric mean sHSP27 361 pg/ml, 95% CI 282, 461, p < 0.05). Participants with few signs of neuropathy (first tertile, NDS ≤2) had significantly higher sHSP27 levels (geometric mean sHSP27 401 pg/ml, 95% CI 310, 520) than participants with many signs (third tertile, NDS ≥7) (geometric mean sHSP27 192 pg/ml, 95% CI 128, 288, p = 0.007). The highest sHSP27 tertile was associated with better nerve function, adjusted for age, sex, statin medication and HbA(1c) (OR 2.51, 95% CI 1.25, 5.05, p < 0.05). CONCLUSIONS/INTERPRETATION High sHSP27 levels were associated with better nerve function and fewer neuropathic signs in NGT, IGT and type 2 diabetes.
Collapse
Affiliation(s)
- K Pourhamidi
- Department of Public Health and Clinical Medicine, Family Medicine, Umeå University, 901 85 Umeå, Sweden.
| | | | | | | |
Collapse
|
11
|
Dokas LA, Malone AM, Williams FE, Nauli SM, Messer WS. Multiple protein kinases determine the phosphorylated state of the small heat shock protein, HSP27, in SH-SY5Y neuroblastoma cells. Neuropharmacology 2011; 61:12-24. [PMID: 21338617 PMCID: PMC3105189 DOI: 10.1016/j.neuropharm.2011.02.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Revised: 01/25/2011] [Accepted: 02/11/2011] [Indexed: 01/21/2023]
Abstract
In SH-SY5Y human neuroblastoma cells, the cholinergic agonist, carbachol, stimulates phosphorylation of the small heat shock protein 27 (HSP27). Carbachol increases phosphorylation of both Ser-82 and Ser-78 while the phorbol ester, phorbol-12, 13-dibutyrate (PDB) affects only Ser-82. Muscarinic receptor activation by carbachol was confirmed by sensitivity of Ser-82 phosphorylation to hyoscyamine with no effect of nicotine or bradykinin. This response to carbachol is partially reduced by inhibition of protein kinase C (PKC) with GF 109203X and p38 mitogen-activated protein kinase (MAPK) with SB 203580. In contrast, phosphorylation produced by PDB is completely reversed by GF 109203X or CID 755673, an inhibitor of PKD. Inhibition of phosphatidylinositol 3-kinase or Akt with LY 294002 or Akti-1/2 stimulates HSP27 phosphorylation while rapamycin, which inhibits mTORC1, does not. The stimulatory effect of Akti-1/2 is reversed by SB 203580 and correlates with increased p38 MAPK phosphorylation. SH-SY5Y cells differentiated with a low concentration of PDB and basic fibroblast growth factor to a more neuronal phenotype retain carbachol-, PDB- and Akti-1/2-responsive HSP27 phosphorylation. Immunofluorescence microscopy confirms increased HSP27 phosphorylation in response to carbachol or PDB. At cell margins, PDB causes f-actin to reorganize forming lamellipodial structures from which phospho-HSP27 is segregated. The resultant phenotypic change in cell morphology is dependent upon PKC, but not PKD, activity. The major conclusion from this study is that the phosphorylated state of HSP27 in SH-SY5Y cells results from integrated signaling involving PKC, p38 MAPK and Akt.
Collapse
Affiliation(s)
- Linda A. Dokas
- Department of Pharmacology, College of Pharmacy, 3000 Arlington Avenue, The University of Toledo, Toledo OH 43614 USA
| | - Amy M. Malone
- Department of Pharmacology, College of Pharmacy, 3000 Arlington Avenue, The University of Toledo, Toledo OH 43614 USA
| | - Frederick E. Williams
- Department of Pharmacology, College of Pharmacy, 3000 Arlington Avenue, The University of Toledo, Toledo OH 43614 USA
| | - Surya M. Nauli
- Department of Pharmacology, College of Pharmacy, 3000 Arlington Avenue, The University of Toledo, Toledo OH 43614 USA
- Department of Medicinal & Biological Chemistry, College of Pharmacy, 3000 Arlington Avenue, The University of Toledo, Toledo OH 43614 USA
| | - William S. Messer
- Department of Pharmacology, College of Pharmacy, 3000 Arlington Avenue, The University of Toledo, Toledo OH 43614 USA
- Department of Medicinal & Biological Chemistry, College of Pharmacy, 3000 Arlington Avenue, The University of Toledo, Toledo OH 43614 USA
| |
Collapse
|
12
|
Liu SP, Ding DC, Wang HJ, Su CY, Lin SZ, Li H, Shyu WC. Nonsenescent Hsp27-Upregulated MSCs Implantation Promotes Neuroplasticity in Stroke Model. Cell Transplant 2010; 19:1261-79. [PMID: 20525429 DOI: 10.3727/096368910x507204] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Cellular senescence induces changes in cellular physiology, morphology, proliferative capacity, and gene expression. Stem cell senescence might be one of the major issues of limited efficacy of stem cell transplantation. In this study, we demonstrated that implantation of human umbilical cord mesenchymal stem cells (hUCMSCs) cultured in human umbilical cord serum (hUCS) significantly enhanced neuroplasticity and angiogenesis in stroke and ischemic limb models. Immunophenotypic analysis indicated that hUCMSCs cultured in hUCS had more small and rapidly self-renewing cells than those expanded in FCS. The main cause of greater senescence in FCS-cultured cells was increased generation of reactive oxygen species (ROS). Proteome profiling showed significantly more senescence-associated vimentin in FCS-cultured hUCMSCs than in hUCS-cultured hUCMSCs. In contrast, there was significant upregulation of heat shock protein 27 (Hsp27) in the hUCS-cultured hUCMSCs. By gene targeting, we found that overexpression of Hsp27 may downregulate vimentin expression through inhibition of the nuclear translocation of p65 (NF-κB signaling). Thus, an interaction between Hsp27 and vimentin may modulate the degree of senescence in hUCS- and FCS-cultured hUCMSCs. In summary, hUCMSCs exhibiting senescence are detrimental to cell engraftment and differentiation in animal models via activation of NF-κB pathway. Human stem cells incubated in hUCS might reduce the senescent process through upregulation of Hsp27 to increase implantation efficiency.
Collapse
Affiliation(s)
- Shih-Ping Liu
- Center for Neuropsychiatry, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Dah-Ching Ding
- Department of Obstetrics and Gynecology, Buddhist Tzu-Chi General Hospital, Hualien, Taiwan
| | - Hsiao-Jung Wang
- Center for Neuropsychiatry, China Medical University Hospital, Taichung, Taiwan
| | - Ching-Yuan Su
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Shinn-Zong Lin
- Center for Neuropsychiatry, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Immunology, China Medical University, Taichung, Taiwan
| | - Hung Li
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Woei-Cherng Shyu
- Center for Neuropsychiatry, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Immunology, China Medical University, Taichung, Taiwan
| |
Collapse
|
13
|
Stetler RA, Gao Y, Signore AP, Cao G, Chen J. HSP27: mechanisms of cellular protection against neuronal injury. Curr Mol Med 2010; 9:863-72. [PMID: 19860665 DOI: 10.2174/156652409789105561] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The heat shock protein (HSP) family has long been associated with a generalized cellular stress response, particularly in terms of recognizing and chaperoning misfolded proteins. While HSPs in general appear to be protective, HSP27 has recently emerged as a particularly potent neuroprotectant in a number of diverse neurological disorders, ranging from ALS to stroke. Although its robust protective effect on a number of insults has been recognized, the mechanisms and regulation of HSP27's protective actions are still undergoing intense investigation. On the basis of recent studies, HSP27 appears to have a dynamic and diverse range of function in cellular survival. This review provides a forum to compare and contrast recent literature exploring the protective mechanism and regulation of HSP27, focusing on neurological disorders in particular, as they represent a range from protein aggregate-associated diseases to acute stress.
Collapse
Affiliation(s)
- R A Stetler
- Department of Neurology, University of Pittsburgh, 507 South Biomedical Science Tower, 200 Lothrop Street, Pittsburgh, PA 15261, USA
| | | | | | | | | |
Collapse
|
14
|
Tucker NR, Ustyugov A, Bryantsev AL, Konkel ME, Shelden EA. Hsp27 is persistently expressed in zebrafish skeletal and cardiac muscle tissues but dispensable for their morphogenesis. Cell Stress Chaperones 2009; 14:521-33. [PMID: 19238587 PMCID: PMC2728285 DOI: 10.1007/s12192-009-0105-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2008] [Revised: 01/29/2009] [Accepted: 02/05/2009] [Indexed: 11/29/2022] Open
Abstract
Constitutive expression of Hsp27 has been demonstrated in vertebrate embryos, especially in developing skeletal and cardiac muscle. Results of several previous studies have indicated that Hsp27 could play a role in the development of these tissues. For example, inhibition of Hsp27 expression has been reported to cause defective development of mammalian myoblasts in vitro and frog embryos in vivo. In contrast, transgenic mice lacking Hsp27 develop normally. Here, we examined the distribution of Hsp27 protein in developing and adult zebrafish and effects of suppressing Hsp27 expression using phosphorodiamidate morpholino oligonucleotides (PMO) on zebrafish development. Consistent with our previous analysis of hsp27 messenger RNA expression, we detected the protein Hsp27 in cardiac, smooth, and skeletal muscle of both embryonic and adult zebrafish. However, embryos lacking detectable Hsp27 after injection of antisense hsp27 PMO exhibited comparable heart beat rates to that of control embryos and cardiac morphology was indistinguishable in the presence or absence of Hsp27. Loss of Hsp27 also had no effect on the structure of the skeletal muscle myotomes in the developing embryo. Finally, embryos injected with antisense hsp27 and scrambled control PMO displayed equal motility. We conclude that Hsp27 is dispensable for zebrafish morphogenesis but could play a role in long-term maintenance of heart and muscle tissues.
Collapse
Affiliation(s)
- Nathan R. Tucker
- School of Molecular Biosciences, Washington State University, Pullman, WA 99164 USA
| | - Alexey Ustyugov
- School of Molecular Biosciences, Washington State University, Pullman, WA 99164 USA
| | - Anton L. Bryantsev
- Department of Biology, University of New Mexico, Albuquerque, NM 87131 USA
| | - Michael E. Konkel
- School of Molecular Biosciences, Washington State University, Pullman, WA 99164 USA
| | - Eric. A. Shelden
- School of Molecular Biosciences, Washington State University, Pullman, WA 99164 USA
- Center for Reproductive Biology, Washington State University, Pullman, WA 99164 USA
| |
Collapse
|
15
|
Sun W, Zhang L, Lu J, Yang G, Laundrie E, Salvi R. Noise exposure-induced enhancement of auditory cortex response and changes in gene expression. Neuroscience 2008; 156:374-80. [PMID: 18713646 DOI: 10.1016/j.neuroscience.2008.07.040] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2008] [Revised: 07/23/2008] [Accepted: 07/23/2008] [Indexed: 10/21/2022]
Abstract
Noise exposure is one of the most common causes of hearing loss. There is growing evidence suggesting that noise-induced peripheral hearing loss can also induce functional changes in the central auditory system. However, the physiological and biological changes in the central auditory system induced by noise exposure are poorly understood. To address these issues, neurophysiological recordings were made from the auditory cortex (AC) of awake rats using chronically implanted electrodes before and after acoustic overstimulation. In addition, focused gene microarrays and quantitative real-time polymerase chain reaction were used to identify changes in gene expression in the AC. Monaural noise exposure (120 dB sound pressure level, 1 h) significantly elevated hearing threshold on the exposed ear and induced a transient enhancement on the AC response amplitude 4 h after the noise exposure recorded from the unexposed ear. This increase of the cortical neural response amplitude was associated with an upregulation of genes encoding heat shock protein (HSP) 27 kDa and 70 kDa after several hours of the noise exposure. These results suggest that noise exposure can induce a fast physiological change in the AC which may be related to the changes of HSP expressions.
Collapse
Affiliation(s)
- W Sun
- Center for Hearing & Deafness, 137 Cary Hall, State University of New York at Buffalo, 3454 Main Street, Buffalo, NY 14214, USA.
| | | | | | | | | | | |
Collapse
|
16
|
Krishnan J, Vannuvel K, Andries M, Waelkens E, Robberecht W, Van Den Bosch L. Over-expression of Hsp27 does not influence disease in the mutant SOD1(G93A) mouse model of amyotrophic lateral sclerosis. J Neurochem 2008; 106:2170-83. [PMID: 18624915 DOI: 10.1111/j.1471-4159.2008.05545.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a chronic, adult-onset neurodegenerative disorder characterized by the selective loss of upper and lower motor neurons, resulting in severe atrophy of muscles and death. Although the exact pathogenic mechanism of mutant superoxide dismutase 1 (SOD1) causing familial ALS is still elusive, toxic protein aggregation leading to insufficiency of chaperones is one of the main hypotheses. In this study, we investigated the effect of over-expressing one of these chaperones, heat shock protein 27 (Hsp27), in ALS. Mice over-expressing the human, mutant SOD1(G93A) were crossed with mice that ubiquitously over-expressed human Hsp27. Even though the single transgenic hHsp27 mice showed protection against spinal cord ischemia, the double transgenic SOD1(G93A)/hHsp27 mice did not live longer, and did not show a significant delay in the onset of disease compared to their SOD1(G93A) littermates. There was no protective effect of hHsp27 over-expression on the motor neurons and on the mutant SOD1 aggregates in the double transgenic SOD1(G93A)/hHsp27 mice. In conclusion, despite the protective action against acute motor neuron injury, Hsp27 alone is not sufficient to protect against the chronic motor neuron injury due to the presence of mutant SOD1.
Collapse
|
17
|
Pituitary adenylate cyclase-activating polypeptide 38-mediated Rin activation requires Src and contributes to the regulation of HSP27 signaling during neuronal differentiation. Mol Cell Biol 2008; 28:4940-51. [PMID: 18541665 DOI: 10.1128/mcb.02193-07] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide 38 (PACAP38) is a potent neuropeptide that acts through G-protein-coupled receptors. While it is well established that PACAP mediates both neurotrophic and neurodevelopmental effects, the signaling cascades that underlie these diverse actions remain incompletely characterized. Here we show that the Ras-related Rin GTP-binding protein, a GTPase that is expressed predominantly in neurons, is regulated by PACAP38 signaling, and loss-of-function analysis demonstrates that Rin makes an essential contribution to PACAP38-mediated pheochromocytoma cell differentiation. Rin is activated following stimulation of both Gsalpha and Gialpha cascades but does not rely upon cyclic AMP (cAMP)-, Ca(2+)-, or Epac-dependent signaling pathways. Instead, Rin is activated in a Src kinase-dependent manner. Surprisingly, Rin knockdown significantly inhibits PACAP38-mediated neurite outgrowth, without affecting mitogen-activated protein kinase signaling cascades. Instead, Rin loss attenuates PACAP38-mediated HSP27 activation by disrupting a cAMP-protein kinase A cascade. RNA interference-mediated HSP27 silencing suppresses both PACAP38- and Rin-mediated neurite outgrowth, while expression of a constitutively active Rin mutant increases both HSP27 protein and phospho-HSP27 levels, supporting a role for Rin-HSP27 signaling in neuronal differentiation. Together, these observations identify an unsuspected role for Rin in neuronal PACAP signaling and establish a novel Galpha-Src-Rin-HSP27 signal transduction pathway as a critical element in PACAP38-mediated neuronal differentiation signaling.
Collapse
|
18
|
Klass MG, Gavrikov V, Krishnamoorthy M, Csete M. Heat shock proteins, endothelin, and peripheral neuronal injury. Neurosci Lett 2008; 433:188-93. [DOI: 10.1016/j.neulet.2008.01.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2007] [Revised: 12/16/2007] [Accepted: 01/03/2008] [Indexed: 11/28/2022]
|
19
|
Hayes K, Sprague S, Guo M, Davis W, Friedman A, Kumar A, Jimenez DF, Ding Y. Forced, not voluntary, exercise effectively induces neuroprotection in stroke. Acta Neuropathol 2008; 115:289-96. [PMID: 18210137 PMCID: PMC2668645 DOI: 10.1007/s00401-008-0340-z] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2007] [Revised: 01/05/2008] [Accepted: 01/06/2008] [Indexed: 11/28/2022]
Abstract
Previous treadmill exercise studies showing neuroprotective effects have raised questions as to whether exercise or the stress related to it may be key etiologic factors. In this study, we examined different exercise regimens (forced and voluntary exercise) and compared them with the effect of stress-only on stroke protection. Adult male Sprague-Dawley rats (n = 65) were randomly assigned to treatment groups for 3 weeks. These groups included control, treadmill exercise, voluntary running wheel exercise, restraint, and electric shock. Levels of the stress hormone, corticosterone, were measured in the different groups using ELISA. Animals from each group were then subjected to stroke induced by a 2-h middle cerebral artery (MCA) occlusion followed by 48-h reperfusion. Infarct volume was determined in each group, while changes in gene expression of stress-induced heat shock proteins (Hsp) 27 and 70 were compared using real-time PCR between voluntary and treadmill exercise groups. The level of corticosterone was significantly higher in both stress (P < 0.05) and treadmill exercise (P < 0.05) groups, but not in the voluntary exercise group. Infarct volume was significantly reduced (P < 0.01) following stroke in rats exercised on a treadmill. However, the amelioration of damage was not duplicated in voluntary exercise, even though running distance in the voluntary exercise group was significantly (P < 0.01) longer than that of the forced exercise group (4,828 vs. 900 m). Furthermore, rats in the electric shock group displayed a significantly increased (P < 0.01) infarct volume. Expression of both Hsp 27 and Hsp 70 mRNA was significantly increased (P < 0.01) in the treadmill exercise group as compared with that in the voluntary exercise group. These results suggest that exercise with a stressful component, rather than either voluntary exercise or stress alone, is better able to reduce infarct volume. This exercise-induced neuroprotection may be attributable to up-regulation of stress-induced heat shock proteins 27 and 70.
Collapse
Affiliation(s)
- Katherine Hayes
- Department of Neurosurgery, The University of Texas Health Science Center at San Antonio, Mail Code 7843, 7703 Floyd Curl Drive, San Antonio, TX 78229-3900 USA
| | - Shane Sprague
- Department of Neurosurgery, The University of Texas Health Science Center at San Antonio, Mail Code 7843, 7703 Floyd Curl Drive, San Antonio, TX 78229-3900 USA
| | - Miao Guo
- Department of Neurosurgery, The University of Texas Health Science Center at San Antonio, Mail Code 7843, 7703 Floyd Curl Drive, San Antonio, TX 78229-3900 USA
| | - William Davis
- Department of Neurosurgery, The University of Texas Health Science Center at San Antonio, Mail Code 7843, 7703 Floyd Curl Drive, San Antonio, TX 78229-3900 USA
| | - Asher Friedman
- Department of Neurosurgery, The University of Texas Health Science Center at San Antonio, Mail Code 7843, 7703 Floyd Curl Drive, San Antonio, TX 78229-3900 USA
| | - Ashwini Kumar
- Department of Neurosurgery, The University of Texas Health Science Center at San Antonio, Mail Code 7843, 7703 Floyd Curl Drive, San Antonio, TX 78229-3900 USA
| | - David F. Jimenez
- Department of Neurosurgery, The University of Texas Health Science Center at San Antonio, Mail Code 7843, 7703 Floyd Curl Drive, San Antonio, TX 78229-3900 USA
| | - Yuchuan Ding
- Department of Neurosurgery, The University of Texas Health Science Center at San Antonio, Mail Code 7843, 7703 Floyd Curl Drive, San Antonio, TX 78229-3900 USA
| |
Collapse
|
20
|
Abstract
Drinking alcohol causes widespread alterations in gene expression that can result in long-term physiological changes. Although many alcohol-responsive genes (ARGs) have been identified, the mechanisms by which alcohol alters transcription are not well understood. To elucidate these mechanisms, we investigated Gabra4, a neuron-specific gene that is rapidly and robustly activated by alcohol (10-60 mM), both in vitro and in vivo. Here we show that alcohol can activate elements of the heat shock pathway in mouse cortical neurons to enhance the expression of Gabra4 and other ARGs. The activation of Gabra4 by alcohol or high temperature is dependent on the binding of heat shock factor 1 (HSF1) to a short downstream DNA sequence, the alcohol response element (ARE). Alcohol and heat stimulate the translocation of HSF1 from the cytoplasm to the nucleus and the induction of HSF1-dependent genes, Hsp70 and Hsp90, in cultured neurons and in the mouse cerebral cortex in vivo. The reduction of HSF1 levels using small interfering RNA prevented the stimulation of Gabra4 and Hsp70 by alcohol and heat shock. Microarray analysis showed that many ARGs contain ARE-like sequences and that some of these genes are also activated by heat shock. We suggest that alcohol activates phylogenetically conserved pathways that involve intermediates in the heat shock cascade and that sequence elements similar to the ARE may mediate some of the changes in gene expression triggered by alcohol intake, which could be important in a variety of pathophysiological responses to alcohol.
Collapse
|
21
|
Raivich G, Makwana M. The making of successful axonal regeneration: Genes, molecules and signal transduction pathways. ACTA ACUST UNITED AC 2007; 53:287-311. [PMID: 17079020 DOI: 10.1016/j.brainresrev.2006.09.005] [Citation(s) in RCA: 131] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2006] [Revised: 09/12/2006] [Accepted: 09/18/2006] [Indexed: 12/16/2022]
Abstract
Unlike its central counterpart, the peripheral nervous system is well known for its comparatively good potential for regeneration following nerve fiber injury. This ability is mirrored by the de novo expression or upregulation of a wide variety of molecules including transcription factors, growth-stimulating substances, cell adhesion molecules, intracellular signaling enzymes and proteins involved in regulating cell-surface cytoskeletal interactions, that promote neurite outgrowth in cultured neurons. However, their role in vivo is less known. Recent studies using neutralizing antibodies, gene inactivation and overexpression techniques have started to shed light on those endogenous molecules that play a key role in axonal outgrowth and the process of successful functional repair in the injured nervous system. The aim of the current review is to provide a summary on this rapidly growing field and the experimental techniques used to define the specific effects of candidate signaling molecules on axonal regeneration in vivo.
Collapse
Affiliation(s)
- Gennadij Raivich
- Perinatal Brain Repair Group, Department of Obstetrics and Gynaecology, University College London, 86-96 Chenies Mews, London, UK.
| | | |
Collapse
|
22
|
Labriola L, Ferreira GB, Montor WR, Demasi MAA, Pimenta DC, Lojudice FH, Genzini T, Goldberg AC, Eliaschewitz FG, Sogayar MC. Prolactin-induced changes in protein expression in human pancreatic islets. Mol Cell Endocrinol 2007; 264:16-27. [PMID: 17095147 DOI: 10.1016/j.mce.2006.10.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2006] [Revised: 10/02/2006] [Accepted: 10/03/2006] [Indexed: 11/26/2022]
Abstract
Ex vivo islet cell culture prior to transplantation appears as an attractive alternative for treatment of type 1 diabetes. Previous results from our laboratory have demonstrated beneficial effects of human prolactin (rhPRL) treatment on human islet primary cultures. In order to probe into the molecular events involved in the intracellular action of rhPRL in these cells, we set out to identify proteins with altered expression levels upon rhPRL cell treatment, using two-dimensional (2D) gel electrophoresis and mass spectrometry (MS). An average of 300 different protein spots were detected, 14 of which were modified upon rhPRL treatment (p<0.01), of which 12 were successfully identified using MS and grouped according to their biological functions. In conclusion, our study provides, for the first time, information about proteins that could be critically involved in PRL's action on human pancreatic islets, and facilitate identification of new and specific targets involved in islet cell function and proliferation.
Collapse
Affiliation(s)
- L Labriola
- Department of Biochemistry, Chemistry Institute, University of Sao Paulo, Av. Prof. Lineu Prestes, 748, Bloco 9 Superior Sala 964, São Paulo 05508-900 SP, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|