1
|
Sangar D, Hill E, Jack K, Batchelor M, Mistry B, Ribes JM, Jackson GS, Mead S, Bieschke J. Syntaxin-6 delays prion protein fibril formation and prolongs the presence of toxic aggregation intermediates. eLife 2024; 13:e83320. [PMID: 39109999 PMCID: PMC11377041 DOI: 10.7554/elife.83320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 07/20/2024] [Indexed: 09/06/2024] Open
Abstract
Prions replicate via the autocatalytic conversion of cellular prion protein (PrPC) into fibrillar assemblies of misfolded PrP. While this process has been extensively studied in vivo and in vitro, non-physiological reaction conditions of fibril formation in vitro have precluded the identification and mechanistic analysis of cellular proteins, which may alter PrP self-assembly and prion replication. Here, we have developed a fibril formation assay for recombinant murine and human PrP (23-231) under near-native conditions (NAA) to study the effect of cellular proteins, which may be risk factors or potential therapeutic targets in prion disease. Genetic screening suggests that variants that increase syntaxin-6 expression in the brain (gene: STX6) are risk factors for sporadic Creutzfeldt-Jakob disease. Analysis of the protein in NAA revealed, counterintuitively, that syntaxin-6 is a potent inhibitor of PrP fibril formation. It significantly delayed the lag phase of fibril formation at highly sub-stoichiometric molar ratios. However, when assessing toxicity of different aggregation time points to primary neurons, syntaxin-6 prolonged the presence of neurotoxic PrP species. Electron microscopy and super-resolution fluorescence microscopy revealed that, instead of highly ordered fibrils, in the presence of syntaxin-6 PrP formed less-ordered aggregates containing syntaxin-6. These data strongly suggest that the protein can directly alter the initial phase of PrP self-assembly and, uniquely, can act as an 'anti-chaperone', which promotes toxic aggregation intermediates by inhibiting fibril formation.
Collapse
Affiliation(s)
- Daljit Sangar
- MRC Prion Unit at UCL, Institute of Prion DiseasesLondonUnited Kingdom
| | - Elizabeth Hill
- MRC Prion Unit at UCL, Institute of Prion DiseasesLondonUnited Kingdom
| | - Kezia Jack
- MRC Prion Unit at UCL, Institute of Prion DiseasesLondonUnited Kingdom
| | - Mark Batchelor
- MRC Prion Unit at UCL, Institute of Prion DiseasesLondonUnited Kingdom
| | - Beenaben Mistry
- MRC Prion Unit at UCL, Institute of Prion DiseasesLondonUnited Kingdom
| | - Juan M Ribes
- MRC Prion Unit at UCL, Institute of Prion DiseasesLondonUnited Kingdom
| | - Graham S Jackson
- MRC Prion Unit at UCL, Institute of Prion DiseasesLondonUnited Kingdom
| | - Simon Mead
- MRC Prion Unit at UCL, Institute of Prion DiseasesLondonUnited Kingdom
| | - Jan Bieschke
- MRC Prion Unit at UCL, Institute of Prion DiseasesLondonUnited Kingdom
| |
Collapse
|
2
|
Wang Q, Zhou Y, Zheng N, Jiang F, Juan C. Identification of hub genes associated with pyroptosis in diabetic nephropathy patients using integrated bioinformatic analysis. Int Urol Nephrol 2024:10.1007/s11255-024-04158-7. [PMID: 39028495 DOI: 10.1007/s11255-024-04158-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 07/12/2024] [Indexed: 07/20/2024]
Abstract
OBJECTIVES To investigate the role of pyroptosis in diabetic nephropathy (DN) and identify potential biomarkers for diagnosis. METHODS We analyzed the GEO dataset GSE96804 to identify differentially expressed genes (DEGs) related to pyroptosis in DN. The CIBERSORT method was used to assess M1 macrophage infiltration in the samples. Using weighted gene co-expression network analysis (WGCNA), we identified gene modules associated with M1 macrophages. The least absolute shrinkage and selection operator (LASSO) method was then applied to screen for key genes. The intersection of key genes identified by LASSO and the gene modules obtained from WGCNA resulted in the identification of ten hub genes as potential biomarkers for DN. RESULTS A total of 366 DEGs were identified, with 310 genes associated with pyroptosis. Increased M1 macrophage infiltration was observed in DN patients. Ten hub genes were identified as potential DN biomarkers: ECM1, LRP2BP, ALKBH7, CDH10, DUSP1, HSPA1A, LPL, NFIL3, PDK4, and TMEM150C. CONCLUSIONS This study highlights the importance of pyroptosis in DN pathophysiology and identifies 10 hub genes as potential biomarkers. These findings may contribute to improved diagnosis and treatment of DN.
Collapse
Affiliation(s)
- Qiuli Wang
- Department of Nephrology, Lianyungang Hospital of Traditional Chinese Medicine, Lianyungang Affiliated Hospital of Nanjing University of Chinese Medicine, Lianyungang, China
- Department of Nephrology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yan Zhou
- Department of Nephrology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Nan Zheng
- Department of Nephrology, Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
| | - Feng Jiang
- Department of Neonatology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China.
| | - Chenxia Juan
- Department of Nephrology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
3
|
Akdag M, van Schijndel V, Sinnige T. Islet amyloid polypeptide tagged with green fluorescent protein localises to mitochondria and forms filamentous aggregates in Caenorhabditis elegans. Biophys Chem 2024; 307:107180. [PMID: 38241827 DOI: 10.1016/j.bpc.2024.107180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/22/2023] [Accepted: 01/12/2024] [Indexed: 01/21/2024]
Abstract
Type 2 diabetes (T2D) is the most common form of diabetes and represents a growing health concern. A characteristic feature of T2D is the aggregation of islet amyloid polypeptide (IAPP), which is thought to be associated with the death of pancreatic β-cells. Inhibiting IAPP aggregation is a promising therapeutic avenue to treat T2D, but the mechanisms of aggregation and toxicity are not yet fully understood. Caenorhabditis elegans is a well-characterised multicellular model organism that has been extensively used to study protein aggregation diseases. In this study, we aimed to develop a simple in vivo model to investigate IAPP aggregation and toxicity based on expression in the C. elegans body wall muscle cells. We show that IAPP tagged with green fluorescent protein (GFP) localises to mitochondria not only in muscle cells but also when expressed in the intestine, in line with previous observations in mouse and human pancreatic β-cells. The IAPP-GFP fusion protein forms solid aggregates, which have a filamentous appearance as seen by electron microscopy. However, the animals expressing IAPP-GFP in the body wall muscle cells do not display a strong motility phenotype, suggesting that the IAPP-GFP aggregates are not considerably toxic. Nevertheless, the mitochondrial localisation and aggregate formation may be useful read-outs to screen for IAPP-solubilizing compounds as a therapeutic strategy for T2D.
Collapse
Affiliation(s)
- Mehmet Akdag
- Membrane Biochemistry and Biophysics, Bijvoet Centre for Biomolecular Research, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
| | - Vera van Schijndel
- Membrane Biochemistry and Biophysics, Bijvoet Centre for Biomolecular Research, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
| | - Tessa Sinnige
- Membrane Biochemistry and Biophysics, Bijvoet Centre for Biomolecular Research, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands.
| |
Collapse
|
4
|
Alfa J, Ben A, Buxaderas E, Akpa P, Hanifah A, Oseni OML, Kenechukwu FC, Mumuni MA, Diaz DD. Development and Evaluation of PEG-gelatin-based Microparticles to Enhance the Oral Delivery of Insulin. Curr Pharm Des 2024; 30:1939-1948. [PMID: 38847248 DOI: 10.2174/0113816128309449240527053640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 05/14/2024] [Indexed: 09/21/2024]
Abstract
BACKGROUND Diabetes mellitus is a global disease identified by hyperglycemia due to defects in insulin secretion, insulin action, or both. OBJECTIVE The main objective of this research was to evaluate the ability of gelatinized Poly(ethylene glycol) (PEG) microparticles to be used as carriers for oral insulin delivery via double emulsion preparation. METHODS Five different batches of the formulation consisting of gelatin:PEG were prepared as follows: 0:1 (W1), 1:0 (W2), 1:1 (W3), 1:3 (W4), and 3:1 (W5). The prepared microparticles (from insulin-loaded batches) had particle sizes ranging from 19.5 ± 0.32-23.9 ± 0.22 μm and encapsulation and loading capacities ranging from 78.8 ± 0.24-88.9 ± 0.95 and 22.2 ± 0.96-29.7 ± 0.86%, respectively. The minimum and maximum in vitro release rates were 8.0 and 66.0%, respectively, for batches W1 and W2 at 8 h. RESULTS Insulin-loaded MPs induced a significant decrease in glucose levels, with a reduction from 100 to 33.35% in batch W5 at 9 h compared to that of subcutaneous insulin (100 to 22.63%). A liver function study showed that the formulation caused no obvious toxicity to the experimental rats. CONCLUSION Gelatinized PEG-based microparticles as insulin delivery systems may open a new window into the development of oral insulin for diabetic treatment.
Collapse
Affiliation(s)
- John Alfa
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Bingham University, Karu, Nasarawa State, Nigeria
| | - Amadi Ben
- Department of Pharmaceutical Technology and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Eduardo Buxaderas
- Instituto Universitario de Bio-Orgánica Antonio González, Astrofísico Francisco Sánchez 2, La Laguna 38206, Tenerife, Spain
- Departamento de Química, Instituto de Química del Sur, INQUISUR (CONICET-UNS), Universidad Nacional del Sur, Avenida Alem 1253, Bahía Blanca 8000, Argentina
| | - Paul Akpa
- Drug Delivery Research Unit, Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Abdulmumin Hanifah
- Department of Medical Laboratory Sciences, Usmanu Danfodiyo University Sokoto, Sokoto, Nigeria
| | - Okolo Martin-Luther Oseni
- Department of Microbiology, Faculty of Natural Sciences, Kogi State University, Ayingba, Kogi State, Nigeria
| | - Franklin C Kenechukwu
- Drug Delivery Research Unit, Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Momoh A Mumuni
- Drug Delivery Research Unit, Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, University of Nigeria, Nsukka, Enugu State, Nigeria
- Department of Microbiology, Faculty of Natural Sciences, Kogi State University, Ayingba, Kogi State, Nigeria
| | - David Diaz Diaz
- Instituto Universitario de Bio-Orgánica Antonio González, Astrofísico Francisco Sánchez 2, La Laguna 38206, Tenerife, Spain
- Departamento de Química Orgánica, Universidad de La Laguna, Avda. Astrofísico Francisco Sánchez 3, La Laguna 38206, Tenerife, Spain
| |
Collapse
|
5
|
St-Louis JL, El Jellas K, Velasco K, Slipp BA, Hu J, Helgeland G, Steine SJ, De Jesus DF, Kulkarni RN, Molven A. Deficiency of the metabolic enzyme SCHAD in pancreatic β-cells promotes amino acid-sensitive hypoglycemia. J Biol Chem 2023; 299:104986. [PMID: 37392854 PMCID: PMC10407745 DOI: 10.1016/j.jbc.2023.104986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 06/02/2023] [Accepted: 06/20/2023] [Indexed: 07/03/2023] Open
Abstract
Congenital hyperinsulinism of infancy (CHI) can be caused by a deficiency of the ubiquitously expressed enzyme short-chain 3-hydroxyacyl-CoA dehydrogenase (SCHAD). To test the hypothesis that SCHAD-CHI arises from a specific defect in pancreatic β-cells, we created genetically engineered β-cell-specific (β-SKO) or hepatocyte-specific (L-SKO) SCHAD knockout mice. While L-SKO mice were normoglycemic, plasma glucose in β-SKO animals was significantly reduced in the random-fed state, after overnight fasting, and following refeeding. The hypoglycemic phenotype was exacerbated when the mice were fed a diet enriched in leucine, glutamine, and alanine. Intraperitoneal injection of these three amino acids led to a rapid elevation in insulin levels in β-SKO mice compared to controls. Consistently, treating isolated β-SKO islets with the amino acid mixture potently enhanced insulin secretion compared to controls in a low-glucose environment. RNA sequencing of β-SKO islets revealed reduced transcription of β-cell identity genes and upregulation of genes involved in oxidative phosphorylation, protein metabolism, and Ca2+ handling. The β-SKO mouse offers a useful model to interrogate the intra-islet heterogeneity of amino acid sensing given the very variable expression levels of SCHAD within different hormonal cells, with high levels in β- and δ-cells and virtually absent α-cell expression. We conclude that the lack of SCHAD protein in β-cells results in a hypoglycemic phenotype characterized by increased sensitivity to amino acid-stimulated insulin secretion and loss of β-cell identity.
Collapse
Affiliation(s)
- Johanna L St-Louis
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, USA; Department of Clinical Medicine, Gade Laboratory for Pathology, University of Bergen, Bergen, Norway
| | - Khadija El Jellas
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, USA; Department of Clinical Medicine, Gade Laboratory for Pathology, University of Bergen, Bergen, Norway
| | - Kelly Velasco
- Department of Clinical Medicine, Gade Laboratory for Pathology, University of Bergen, Bergen, Norway
| | - Brittany A Slipp
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, USA
| | - Jiang Hu
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, USA
| | - Geir Helgeland
- Department of Clinical Medicine, Gade Laboratory for Pathology, University of Bergen, Bergen, Norway
| | - Solrun J Steine
- Department of Clinical Medicine, Gade Laboratory for Pathology, University of Bergen, Bergen, Norway
| | - Dario F De Jesus
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, USA; Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA; Harvard Stem Cell Institute, Boston, USA
| | - Rohit N Kulkarni
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, USA; Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA; Harvard Stem Cell Institute, Boston, USA
| | - Anders Molven
- Department of Clinical Medicine, Gade Laboratory for Pathology, University of Bergen, Bergen, Norway; Department of Pathology, Haukeland University Hospital, Bergen, Norway; Section for Cancer Genomics, Haukeland University Hospital, Bergen, Norway.
| |
Collapse
|
6
|
Zhou Z, Li S, Gong X. Polydopamine Nanoparticles-Based Photothermal Effect Against Adhesion Formation in a Rat Model of Achilles Tendon Laceration Repair. Int J Nanomedicine 2023; 18:1765-1776. [PMID: 37038441 PMCID: PMC10082603 DOI: 10.2147/ijn.s393454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 03/21/2023] [Indexed: 04/05/2023] Open
Abstract
Background Adhesion formation after tendon surgery is a major obstacle to repair of tendon ruptures, and there is still no effective clinical anti-adhesion method. Myofibroblasts expressing α-smooth muscle actin (α-SMA) play a crucial role in adhered fibrous tissue. Heat shock protein (Hsp) 72 can selectively prevent the activation of c-Jun N-terminal kinase (JNK), which mediates the conversion from fibroblasts to myofibroblasts. The purpose of this study was to investigate for the first time whether polydopamine nanoparticles (PDA NPs)-based photothermal effect would attenuate adhesion formation in a rat model of Achilles tendon laceration repair. Materials and Methods Forty-five adult male Sprague-Dawley rats were randomly assigned to the photothermal group, the control group and the PDA NPs group (n = 15 per group). The primary outcome measure was the adhesion scores at two weeks after surgery according to the grading of Tang et al. The secondary outcomes included the expressions of Hsp 72, JNK, phosphorylated JNK and α-SMA, which were measured by immunohistochemistry or Western blot. Results The average adhesion score was significantly lower in the photothermal group (4.25 ± 0.21) than that in the control group (5.29 ± 0.12) (p = 0.005) and the PDA NPs group (5.29 ± 0.20) (p = 0.005). Relative to the control group and PDA NPs group, Hsp 72 in the photothermal group was significantly increased whereas α-SMA and p-JNK was significantly decreased, but JNK was not found to be different across the three groups. Conclusion The photothermal effect produced by PDA NPs could reduce tendon adhesion formation in rats by inhibiting myocyte fibrosis, which may have potential in developing endogenous heating for postsurgical tissue adhesions.
Collapse
Affiliation(s)
- Zekun Zhou
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, 130021, People’s Republic of China
- Jilin Province Key Laboratory on Tissue Repair, Reconstruction and Regeneration, The First Hospital of Jilin University, Changchun, 130021, People’s Republic of China
| | - Shaoyan Li
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, 130021, People’s Republic of China
- Jilin Province Key Laboratory on Tissue Repair, Reconstruction and Regeneration, The First Hospital of Jilin University, Changchun, 130021, People’s Republic of China
| | - Xu Gong
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, 130021, People’s Republic of China
- Jilin Province Key Laboratory on Tissue Repair, Reconstruction and Regeneration, The First Hospital of Jilin University, Changchun, 130021, People’s Republic of China
- Correspondence: Xu Gong, Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, 130021, People’s Republic of China, Tel +86 13944099151, Email
| |
Collapse
|
7
|
Molecular Mechanisms of Amylin Turnover, Misfolding and Toxicity in the Pancreas. Molecules 2022; 27:molecules27031021. [PMID: 35164285 PMCID: PMC8838401 DOI: 10.3390/molecules27031021] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/24/2022] [Accepted: 01/29/2022] [Indexed: 12/13/2022] Open
Abstract
Amyloidosis is a common pathological event in which proteins self-assemble into misfolded soluble and insoluble molecular forms, oligomers and fibrils that are often toxic to cells. Notably, aggregation-prone human islet amyloid polypeptide (hIAPP), or amylin, is a pancreatic hormone linked to islet β-cells demise in diabetics. The unifying mechanism by which amyloid proteins, including hIAPP, aggregate and kill cells is still matter of debate. The pathology of type-2 diabetes mellitus (T2DM) is characterized by extracellular and intracellular accumulation of toxic hIAPP species, soluble oligomers and insoluble fibrils in pancreatic human islets, eventually leading to loss of β-cell mass. This review focuses on molecular, biochemical and cell-biology studies exploring molecular mechanisms of hIAPP synthesis, trafficking and degradation in the pancreas. In addition to hIAPP turnover, the dynamics and the mechanisms of IAPP–membrane interactions; hIAPP aggregation and toxicity in vitro and in situ; and the regulatory role of diabetic factors, such as lipids and cholesterol, in these processes are also discussed.
Collapse
|
8
|
Marmentini C, Branco RCS, Boschero AC, Kurauti MA. Islet amyloid toxicity: From genesis to counteracting mechanisms. J Cell Physiol 2021; 237:1119-1142. [PMID: 34636428 DOI: 10.1002/jcp.30600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 09/09/2021] [Accepted: 10/01/2021] [Indexed: 11/11/2022]
Abstract
Islet amyloid polypeptide (IAPP or amylin) is a hormone co-secreted with insulin by pancreatic β-cells and is the major component of islet amyloid. Islet amyloid is found in the pancreas of patients with type 2 diabetes (T2D) and may be involved in β-cell dysfunction and death, observed in this disease. Thus, investigating the aspects related to amyloid formation is relevant to the development of strategies towards β-cell protection. In this sense, IAPP misprocessing, IAPP overproduction, and disturbances in intra- and extracellular environments seem to be decisive for IAPP to form islet amyloid. Islet amyloid toxicity in β-cells may be triggered in intra- and/or extracellular sites by membrane damage, endoplasmic reticulum stress, autophagy disruption, mitochondrial dysfunction, inflammation, and apoptosis. Importantly, different approaches have been suggested to prevent islet amyloid cytotoxicity, from inhibition of IAPP aggregation to attenuation of cell death mechanisms. Such approaches have improved β-cell function and prevented the development of hyperglycemia in animals. Therefore, counteracting islet amyloid may be a promising therapy for T2D treatment.
Collapse
Affiliation(s)
- Carine Marmentini
- Laboratory of Endocrine Pancreas and Metabolism, Obesity and Comorbidities Research Center (OCRC), University of Campinas (UNICAMP), Campinas, Brazil
| | - Renato C S Branco
- Laboratory of Endocrine Pancreas and Metabolism, Obesity and Comorbidities Research Center (OCRC), University of Campinas (UNICAMP), Campinas, Brazil
| | - Antonio C Boschero
- Laboratory of Endocrine Pancreas and Metabolism, Obesity and Comorbidities Research Center (OCRC), University of Campinas (UNICAMP), Campinas, Brazil
| | - Mirian A Kurauti
- Laboratory of Endocrine Pancreas and Metabolism, Obesity and Comorbidities Research Center (OCRC), University of Campinas (UNICAMP), Campinas, Brazil.,Department of Physiological Sciences, Biological Sciences Center, State University of Maringa (UEM), Maringa, Brazil
| |
Collapse
|
9
|
Ren X, Zhou C, Lu Y, Ma F, Fan Y, Wang C. Single-cell RNA-seq reveals invasive trajectory and determines cancer stem cell-related prognostic genes in pancreatic cancer. Bioengineered 2021; 12:5056-5068. [PMID: 34474642 PMCID: PMC8806718 DOI: 10.1080/21655979.2021.1962484] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pancreatic duct adenocarcinoma (PDAC) is an aggressive and lethal malignancy. Pancreatic cancer stem cells (PCSCs) are assumed to contribute to the initiation and invasion of PDAC. In this study, we performed single-cell RNA sequencing (scRNA-seq) analysis of PDAC tumor samples from patients and control pancreas tissues to reveal the transformation process of cancer stem cell (CSC)-like ductal cells into ductal cells with invasive potential and we screened out CSC-related genes (CRGs). Subsequently, we applied LASSO and Cox regression models to identify five CRGs with potential prognostic values and constructed a risk prognostic model using the Cancer Genome Atlas datasets. The risk models were verified using Gene Expression Omnibus datasets. Patients in the high-risk group had a significantly poor overall survival (Pvalue<0.0001), as illustrated by the Kaplan-Meier survival curve, and the area under the curve confirmed the accuracy of predictions by our risk model. Tumor mutation burden variations were used to further explore the differences between the two risk cohorts. In addition, the Human Protein Atlas was used to investigate the protein expression of five hub CRGs. In brief, we utilized scRNA-seq to reveal the invasive trajectory of ductal cells and identified crucial CRGs in PDAC, which may help predict patient survival and provide potential clinical therapeutic targets against CSCs.
Collapse
Affiliation(s)
- Xuechen Ren
- Lanzhou University Second Hospital, Lanzhou, GS, China
| | | | - Yu Lu
- Lanzhou University Second Hospital, Lanzhou, GS, China
| | - Fulin Ma
- Lanzhou University Second Hospital, Lanzhou, GS, China
| | - Yong Fan
- Lanzhou University Second Hospital, Lanzhou, GS, China
| | - Chen Wang
- Lanzhou University Second Hospital, Lanzhou, GS, China
| |
Collapse
|
10
|
Diane A, Abunada H, Khattab N, Moin ASM, Butler AE, Dehbi M. Role of the DNAJ/HSP40 family in the pathogenesis of insulin resistance and type 2 diabetes. Ageing Res Rev 2021; 67:101313. [PMID: 33676026 DOI: 10.1016/j.arr.2021.101313] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 02/22/2021] [Accepted: 02/28/2021] [Indexed: 12/13/2022]
Abstract
Insulin resistance (IR) underpins a wide range of metabolic disorders including type 2 diabetes (T2D), metabolic syndrome and cardiovascular diseases. IR is characterized by a marked reduction in the magnitude and/or delayed onset of insulin to stimulate glucose disposal. This condition is due to defects in one or several intracellular intermediates of the insulin signaling cascade, ranging from insulin receptor substrate (IRS) inactivation to reduced glucose phosphorylation and oxidation. Genetic predisposition, as well as other precipitating factors such as aging, obesity, and sedentary lifestyles are among the risk factors underlying the pathogenesis of IR and its subsequent progression to T2D. One of the cardinal hallmarks of T2D is the impairment of the heat shock response (HSR). Human and animal studies provided compelling evidence of reduced expression of several components of the HSR (i.e. Heat shock proteins or HSPs) in diabetic samples in a manner that correlates with the degree of IR. Interventions that induce the HSR, irrespective of the means to achieve it, proved their effectiveness in enhancing insulin sensitivity and improving glycemic index. However, most of these studies have been focused on HSP70 family. In this review, we will focus on the novel role of DNAJ/HSP40 cochaperone family in metabolic diseases associated with IR.
Collapse
|
11
|
Chilukoti N, Sil TB, Sahoo B, Deepa S, Cherakara S, Maddheshiya M, Garai K. Hsp70 Inhibits Aggregation of IAPP by Binding to the Heterogeneous Prenucleation Oligomers. Biophys J 2021; 120:476-488. [PMID: 33417920 PMCID: PMC7895988 DOI: 10.1016/j.bpj.2020.12.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 12/08/2020] [Accepted: 12/10/2020] [Indexed: 12/11/2022] Open
Abstract
Molecular chaperone Hsp70 plays important roles in the pathology of amyloid diseases by inhibiting aberrant aggregation of proteins. However, the biophysical mechanism of the interaction of Hsp70 with the intrinsically disordered proteins (IDPs) is unclear. Here, we report that Hsp70 inhibits aggregation of islet amyloid polypeptide (IAPP) at substoichiometric concentrations under diverse solution conditions, including in the absence of ATP. The inhibitory effect is strongest if Hsp70 is added in the beginning of aggregation but progressively less if added later, indicating a role for Hsp70 in preventing nucleation of IAPP. However, ensemble measurement of the binding affinity suggests poor interactions between Hsp70 and IAPP. Therefore, we hypothesize that the interaction must involve a rare species (e.g., the oligomeric intermediates of IAPP). Size exclusion chromatography and field flow fractionation are then used to fractionate the constituent species. Multiangle light scattering and fluorescence correlation spectroscopy measurements indicate that the dominant fraction in size exclusion chromatography contains a few nanomolar Hsp70-IAPP complexes amid several μmoles of free Hsp70. Using single-particle two-color coincidence detection measurements, we detected a minor fraction that exhibits fluorescence bursts arising from heterogeneous oligomeric complexes of IAPP and Hsp70. Taken together, our results indicate that Hsp70 interacts poorly with the monomers but strongly with oligomers of IAPP. This is likely a generic feature of the interactions of Hsp70 chaperones with the amyloidogenic IDPs. Whereas high-affinity interactions with the oligomers prevent aberrant aggregation, poor interaction with the monomers averts interference with the physiological functions of the IDPs.
Collapse
Affiliation(s)
- Neeraja Chilukoti
- Tata Institute of Fundamental Research, Serilingampally, Hyderabad, India
| | - Timir Baran Sil
- Tata Institute of Fundamental Research, Serilingampally, Hyderabad, India
| | - Bankanidhi Sahoo
- Tata Institute of Fundamental Research, Serilingampally, Hyderabad, India
| | - S Deepa
- Tata Institute of Fundamental Research, Serilingampally, Hyderabad, India
| | | | - Mithun Maddheshiya
- Tata Institute of Fundamental Research, Serilingampally, Hyderabad, India
| | - Kanchan Garai
- Tata Institute of Fundamental Research, Serilingampally, Hyderabad, India.
| |
Collapse
|
12
|
Piñeros AR, Gao H, Wu W, Liu Y, Tersey SA, Mirmira RG. Single-Cell Transcriptional Profiling of Mouse Islets Following Short-Term Obesogenic Dietary Intervention. Metabolites 2020; 10:metabo10120513. [PMID: 33353164 PMCID: PMC7765825 DOI: 10.3390/metabo10120513] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/14/2020] [Accepted: 12/17/2020] [Indexed: 12/22/2022] Open
Abstract
Obesity is closely associated with adipose tissue inflammation and insulin resistance. Dysglycemia and type 2 diabetes results when islet β cells fail to maintain appropriate insulin secretion in the face of insulin resistance. To clarify the early transcriptional events leading to β-cell failure in the setting of obesity, we fed male C57BL/6J mice an obesogenic, high-fat diet (60% kcal from fat) or a control diet (10% kcal from fat) for one week, and islets from these mice (from four high-fat- and three control-fed mice) were subjected to single-cell RNA sequencing (sc-RNAseq) analysis. Islet endocrine cell types (α cells, β cells, δ cells, PP cells) and other resident cell types (macrophages, T cells) were annotated by transcript profiles and visualized using Uniform Manifold Approximation and Projection for Dimension Reduction (UMAP) plots. UMAP analysis revealed distinct cell clusters (11 for β cells, 5 for α cells, 3 for δ cells, PP cells, ductal cells, endothelial cells), emphasizing the heterogeneity of cell populations in the islet. Collectively, the clusters containing the majority of β cells showed the fewest gene expression changes, whereas clusters harboring the minority of β cells showed the most changes. We identified that distinct β-cell clusters downregulate genes associated with the endoplasmic reticulum stress response and upregulate genes associated with insulin secretion, whereas others upregulate genes that impair insulin secretion, cell proliferation, and cell survival. Moreover, all β-cell clusters negatively regulate genes associated with immune response activation. Glucagon-producing α cells exhibited patterns similar to β cells but, again, in clusters containing the minority of α cells. Our data indicate that an early transcriptional response in islets to an obesogenic diet reflects an attempt by distinct populations of β cells to augment or impair cellular function and/or reduce inflammatory responses as possible harbingers of ensuing insulin resistance.
Collapse
Affiliation(s)
- Annie R. Piñeros
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.R.P.); (W.W.)
| | - Hongyu Gao
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (H.G.); (Y.L.)
| | - Wenting Wu
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.R.P.); (W.W.)
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (H.G.); (Y.L.)
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (H.G.); (Y.L.)
| | - Sarah A. Tersey
- Kolver Diabetes Center and Department of Medicine, The University of Chicago, Chicago, IL 60637, USA;
| | - Raghavendra G. Mirmira
- Kolver Diabetes Center and Department of Medicine, The University of Chicago, Chicago, IL 60637, USA;
- Correspondence:
| |
Collapse
|
13
|
Madhu D, Khadir A, Hammad M, Kavalakatt S, Dehbi M, Al-Mulla F, Abubaker J, Tiss A. The GLP-1 analog exendin-4 modulates HSP72 expression and ERK1/2 activity in BTC6 mouse pancreatic cells. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2020; 1868:140426. [DOI: 10.1016/j.bbapap.2020.140426] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 03/09/2020] [Accepted: 04/02/2020] [Indexed: 12/25/2022]
|
14
|
Wentink A, Nussbaum-Krammer C, Bukau B. Modulation of Amyloid States by Molecular Chaperones. Cold Spring Harb Perspect Biol 2019; 11:a033969. [PMID: 30755450 PMCID: PMC6601462 DOI: 10.1101/cshperspect.a033969] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Aberrant protein aggregation is a defining feature of most neurodegenerative diseases. During pathological aggregation, key proteins transition from their native state to alternative conformations, which are prone to oligomerize into highly ordered fibrillar states. As part of the cellular quality control machinery, molecular chaperones can intervene at many stages of the aggregation process to inhibit or reverse aberrant protein aggregation or counteract the toxicity associated with amyloid species. Although the action of chaperones is considered cytoprotective, essential housekeeping functions can be hijacked for the propagation and spreading of protein aggregates, suggesting the cellular protein quality control system constitutes a double-edged sword in neurodegeneration. Here, we discuss the various mechanisms used by chaperones to influence protein aggregation into amyloid fibrils to understand how the interplay of these activities produces specific cellular outcomes and to define mechanisms that may be targeted by pharmacological agents for the treatment of neurodegenerative conditions.
Collapse
Affiliation(s)
- Anne Wentink
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, D-69120 Heidelberg, Germany
| | - Carmen Nussbaum-Krammer
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, D-69120 Heidelberg, Germany
| | - Bernd Bukau
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, D-69120 Heidelberg, Germany
| |
Collapse
|
15
|
Aldras Y, Singh S, Bode K, Bhowmick DC, Jeremic A, O'Halloran DM. An inducible model of human amylin overexpression reveals diverse transcriptional changes. Neurosci Lett 2019; 704:212-219. [PMID: 30974231 PMCID: PMC6594890 DOI: 10.1016/j.neulet.2019.04.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 04/04/2019] [Accepted: 04/07/2019] [Indexed: 12/31/2022]
Abstract
Human Islet Amyloid Polypeptide or amylin is a neuroendocrine peptide with important endocrine and paracrine functions. Excessive production and accumulation of human amylin in the pancreas can lead to its aggregation and apoptosis of islet β-cells. Amylin has been shown to function within the central nervous system to decrease food intake, and more recently, it has been revealed that amylin is directly transcribed from neurons of the central nervous system, including the hypothalamus, arcuate nucleus, medial preoptic area, and nucleus accumbens. These findings alter the current model of how amylin targets the nervous system, and as a result may lead to obesity and type II diabetes mellitus. Here we set out to use Caenorhabditis elegans as an inducible in vivo model system to study the effects of amylin overexpression in tissues that include the nervous system. We profiled the transcriptional changes in transgenic animals expressing human amylin through RNA-seq. Using this genome-wide approach our results revealed for the first time that expression of human amylin in tissues including the nervous system induce diverse physiological responses in various signaling pathways. From our characterization of transgenic C. elegans animals expressing human amylin, we also observed specific defects in neural developmental programs as well as sensory behavior. Taken together, our data demonstrate the utility of using C. elegans as a valuable in vivo model to study human amylin toxicity.
Collapse
Affiliation(s)
- Yoseph Aldras
- Department of Biological Sciences, The George Washington University, Science and Engineering Hall 6000, 800 22nd St. N.W., Washington DC, 20052, USA; Institute for Neuroscience, The George Washington University, 636 Ross Hall, 2300 I St. N.W. Washington DC, 20052, USA
| | - Sanghamitra Singh
- Department of Biological Sciences, The George Washington University, Science and Engineering Hall 6000, 800 22nd St. N.W., Washington DC, 20052, USA
| | - Katrin Bode
- Department of Biological Sciences, The George Washington University, Science and Engineering Hall 6000, 800 22nd St. N.W., Washington DC, 20052, USA; Institute for Neuroscience, The George Washington University, 636 Ross Hall, 2300 I St. N.W. Washington DC, 20052, USA
| | - Diti Chatterjee Bhowmick
- Department of Biological Sciences, The George Washington University, Science and Engineering Hall 6000, 800 22nd St. N.W., Washington DC, 20052, USA
| | - Aleksandar Jeremic
- Department of Biological Sciences, The George Washington University, Science and Engineering Hall 6000, 800 22nd St. N.W., Washington DC, 20052, USA
| | - Damien M O'Halloran
- Department of Biological Sciences, The George Washington University, Science and Engineering Hall 6000, 800 22nd St. N.W., Washington DC, 20052, USA; Institute for Neuroscience, The George Washington University, 636 Ross Hall, 2300 I St. N.W. Washington DC, 20052, USA.
| |
Collapse
|
16
|
Skórzyńska-Dziduszko KE, Kimber-Trojnar Ż, Patro-Małysza J, Stenzel-Bembenek A, Oleszczuk J, Leszczyńska-Gorzelak B. Heat Shock Proteins as a Potential Therapeutic Target in the Treatment of Gestational Diabetes Mellitus: What We Know so Far. Int J Mol Sci 2018; 19:ijms19103205. [PMID: 30336561 PMCID: PMC6213996 DOI: 10.3390/ijms19103205] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 10/14/2018] [Accepted: 10/15/2018] [Indexed: 12/16/2022] Open
Abstract
Gestational diabetes mellitus (GDM) is a complex condition that involves a variety of pathological mechanisms, including pancreatic β-cell failure, insulin resistance, and inflammation. There is an increasing body of literature suggesting that these interrelated phenomena may arise from the common mechanism of endoplasmic reticulum (ER) stress. Both obesity-associated nutrient excess and hyperglycemia disturb ER function in protein folding and transport. This results in the accumulation of polypeptides in the ER lumen and impairs insulin secretion and signaling. Exercise elicits metabolic adaptive responses, which may help to restore normal chaperone expression in insulin-resistant tissues. Pharmacological induction of chaperones, mimicking the metabolic effect of exercise, is a promising therapeutic tool for preventing GDM by maintaining the body's natural stress response. Metformin, a commonly used diabetes medication, has recently been identified as a modulator of ER-stress-associated inflammation. The results of recent studies suggest the potential use of chemical ER chaperones and antioxidant vitamins as therapeutic interventions that can prevent glucose-induced ER stress in GDM placentas. In this review, we discuss whether chaperones may significantly contribute to the pathogenesis of GDM, as well as whether they can be a potential therapeutic target in GDM treatment.
Collapse
Affiliation(s)
| | - Żaneta Kimber-Trojnar
- Department of Obstetrics and Perinatology, Medical University of Lublin, K. Jaczewskiego 8 Street, 20-954 Lublin, Poland.
| | - Jolanta Patro-Małysza
- Department of Obstetrics and Perinatology, Medical University of Lublin, K. Jaczewskiego 8 Street, 20-954 Lublin, Poland.
| | - Agnieszka Stenzel-Bembenek
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, W. Chodźki 1 Street, 20-093 Lublin, Poland.
| | - Jan Oleszczuk
- Department of Obstetrics and Perinatology, Medical University of Lublin, K. Jaczewskiego 8 Street, 20-954 Lublin, Poland.
| | - Bożena Leszczyńska-Gorzelak
- Department of Obstetrics and Perinatology, Medical University of Lublin, K. Jaczewskiego 8 Street, 20-954 Lublin, Poland.
| |
Collapse
|
17
|
Song M, Wang H, Chen K, Zhang S, Yu L, Elshazly EH, Ke L, Gong R. Oral insulin delivery by carboxymethyl-β-cyclodextrin-grafted chitosan nanoparticles for improving diabetic treatment. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:S774-S782. [DOI: 10.1080/21691401.2018.1511575] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Mingming Song
- College of Life Science, Anhui Normal University, Wuhu, P.R. China
- School of Life Science and Biotechnology, China Pharmaceutical University, Nanjing, P.R. China
| | - Hui Wang
- College of Life Science, Anhui Normal University, Wuhu, P.R. China
| | - Kuanmin Chen
- College of Life Science, Anhui Normal University, Wuhu, P.R. China
| | - Song Zhang
- College of Life Science, Anhui Normal University, Wuhu, P.R. China
| | - Lizhen Yu
- College of Life Science, Anhui Normal University, Wuhu, P.R. China
- School of Pharmacy, Wannan Medical College, Wuhu, P.R. China
| | - Ezzat H. Elshazly
- College of Life Science, Anhui Normal University, Wuhu, P.R. China
- Department of Botany and Microbiology, Faculty of Science, Al Azhar University, Assiut, Egypt
| | - Lixia Ke
- College of Life Science, Anhui Normal University, Wuhu, P.R. China
| | - Renmin Gong
- College of Life Science, Anhui Normal University, Wuhu, P.R. China
| |
Collapse
|
18
|
Weber SC. Sequence-encoded material properties dictate the structure and function of nuclear bodies. Curr Opin Cell Biol 2017; 46:62-71. [PMID: 28343140 DOI: 10.1016/j.ceb.2017.03.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 02/13/2017] [Accepted: 03/07/2017] [Indexed: 12/21/2022]
Abstract
Concomitant with packaging the genome, the cell nucleus must also spatially organize the nucleoplasm. This complex mixture of proteins and nucleic acids partitions into a variety of phase-separated, membraneless organelles called nuclear bodies. Significant progress has been made in understanding the relationship between the material properties of nuclear bodies and their structural and functional consequences. Furthermore, the molecular basis of these condensed phases is beginning to emerge. Here, I review the latest work in this exciting field, highlighting recent advances and new challenges.
Collapse
Affiliation(s)
- Stephanie C Weber
- Department of Biology, McGill University, Montreal, QC H3A 1B1, Canada.
| |
Collapse
|
19
|
da Luz CM, Boyles MSP, Falagan-Lotsch P, Pereira MR, Tutumi HR, de Oliveira Santos E, Martins NB, Himly M, Sommer A, Foissner I, Duschl A, Granjeiro JM, Leite PEC. Poly-lactic acid nanoparticles (PLA-NP) promote physiological modifications in lung epithelial cells and are internalized by clathrin-coated pits and lipid rafts. J Nanobiotechnology 2017; 15:11. [PMID: 28143572 PMCID: PMC5282631 DOI: 10.1186/s12951-016-0238-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 12/03/2016] [Indexed: 02/07/2023] Open
Abstract
Background Poly-lactic acid nanoparticles (PLA-NP) are a type of polymeric NP, frequently used as nanomedicines, which have advantages over metallic NP such as the ability to maintain therapeutic drug levels for sustained periods of time. Despite PLA-NP being considered biocompatible, data concerning alterations in cellular physiology are scarce. Methods We conducted an extensive evaluation of PLA-NP biocompatibility in human lung epithelial A549 cells using high throughput screening and more complex methodologies. These included measurements of cytotoxicity, cell viability, immunomodulatory potential, and effects upon the cells’ proteome. We used non- and green-fluorescent PLA-NP with 63 and 66 nm diameters, respectively. Cells were exposed with concentrations of 2, 20, 100 and 200 µg/mL, for 24, 48 and 72 h, in most experiments. Moreover, possible endocytic mechanisms of internalization of PLA-NP were investigated, such as those involving caveolae, lipid rafts, macropinocytosis and clathrin-coated pits. Results Cell viability and proliferation were not altered in response to PLA-NP. Multiplex analysis of secreted mediators revealed a low-level reduction of IL-12p70 and vascular epidermal growth factor (VEGF) in response to PLA-NP, while all other mediators assessed were unaffected. However, changes to the cells’ proteome were observed in response to PLA-NP, and, additionally, the cellular stress marker miR155 was found to reduce. In dual exposures of staurosporine (STS) with PLA-NP, PLA-NP enhanced susceptibility to STS-induced cell death. Finally, PLA-NP were rapidly internalized in association with clathrin-coated pits, and, to a lesser extent, with lipid rafts. Conclusions These data demonstrate that PLA-NP are internalized and, in general, tolerated by A549 cells, with no cytotoxicity and no secretion of pro-inflammatory mediators. However, PLA-NP exposure may induce modification of biological functions of A549 cells, which should be considered when designing drug delivery systems. Moreover, the pathways of PLA-NP internalization we detected could contribute to the improvement of selective uptake strategies. Electronic supplementary material The online version of this article (doi:10.1186/s12951-016-0238-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Camila Macedo da Luz
- Laboratory of Bioengineering and in Vitro Toxicology, Directory of Metrology Applied to Life Sciences (Dimav), National Institute of Metrology Quality and Technology (INMETRO), Duque De Caxias, RJ, Brazil
| | - Matthew Samuel Powys Boyles
- Department of Molecular Biology, University of Salzburg, Salzburg, Austria.,Heriot-Watt University, Edinburg, UK
| | - Priscila Falagan-Lotsch
- Laboratory of Bioengineering and in Vitro Toxicology, Directory of Metrology Applied to Life Sciences (Dimav), National Institute of Metrology Quality and Technology (INMETRO), Duque De Caxias, RJ, Brazil
| | - Mariana Rodrigues Pereira
- Laboratory of Chemical Signaling in Nervous System, Biology Institute, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Henrique Rudolf Tutumi
- Laboratory of Bioengineering and in Vitro Toxicology, Directory of Metrology Applied to Life Sciences (Dimav), National Institute of Metrology Quality and Technology (INMETRO), Duque De Caxias, RJ, Brazil
| | - Eidy de Oliveira Santos
- Laboratory of Bioengineering and in Vitro Toxicology, Directory of Metrology Applied to Life Sciences (Dimav), National Institute of Metrology Quality and Technology (INMETRO), Duque De Caxias, RJ, Brazil.,Laboratory of Biochemistry, State University Center of West Zone (UEZO), Rio de Janeiro, RJ, Brazil
| | - Nathalia Balthazar Martins
- Laboratory of Bioengineering and in Vitro Toxicology, Directory of Metrology Applied to Life Sciences (Dimav), National Institute of Metrology Quality and Technology (INMETRO), Duque De Caxias, RJ, Brazil
| | - Martin Himly
- Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | - Aniela Sommer
- Department of Cell Biology, University of Salzburg, Salzburg, Austria
| | - Ilse Foissner
- Department of Cell Biology, University of Salzburg, Salzburg, Austria
| | - Albert Duschl
- Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | - José Mauro Granjeiro
- Laboratory of Bioengineering and in Vitro Toxicology, Directory of Metrology Applied to Life Sciences (Dimav), National Institute of Metrology Quality and Technology (INMETRO), Duque De Caxias, RJ, Brazil.,Dental School, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Paulo Emílio Corrêa Leite
- Laboratory of Bioengineering and in Vitro Toxicology, Directory of Metrology Applied to Life Sciences (Dimav), National Institute of Metrology Quality and Technology (INMETRO), Duque De Caxias, RJ, Brazil. .,, Av. Nossa Senhora das Gracas 50, LABET - Dimav, Predio 27, Duque de Caxias, Xerem, Rio de Janeiro, 25250-020, Brazil.
| |
Collapse
|
20
|
Guo J, Sun W, Li L, Liu F, Lu W. Brazilin inhibits fibrillogenesis of human islet amyloid polypeptide, disassembles mature fibrils, and alleviates cytotoxicity. RSC Adv 2017. [DOI: 10.1039/c7ra05742c] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Inhibitory effect of brazilin on the fibrillogenesis of hIAPP was explored using biochemical, biophysical, cytobiological and molecular simulation experiments. Brazilin was a potential compound for therapeutic treatment of type II diabetes mellitus.
Collapse
Affiliation(s)
- Jingjing Guo
- Department of Biochemical Engineering
- Key Laboratory of Systems Bioengineering of Ministry of Education
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300072
| | - Wanqi Sun
- Department of Chemical and Biological Engineering
- The University of Alabama
- Tuscaloosa
- USA
| | - Li Li
- College of Marine and Environmental Sciences
- Tianjin University of Science & Technology
- Tianjin 300457
- P. R. China
| | - Fufeng Liu
- Department of Biochemical Engineering
- Key Laboratory of Systems Bioengineering of Ministry of Education
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300072
| | - Wenyu Lu
- Department of Biochemical Engineering
- Key Laboratory of Systems Bioengineering of Ministry of Education
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300072
| |
Collapse
|
21
|
Su KH, Dai C. Metabolic control of the proteotoxic stress response: implications in diabetes mellitus and neurodegenerative disorders. Cell Mol Life Sci 2016; 73:4231-4248. [PMID: 27289378 PMCID: PMC5599143 DOI: 10.1007/s00018-016-2291-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Revised: 05/13/2016] [Accepted: 06/07/2016] [Indexed: 12/12/2022]
Abstract
Proteome homeostasis, or proteostasis, is essential to maintain cellular fitness and its disturbance is associated with a broad range of human health conditions and diseases. Cells are constantly challenged by various extrinsic and intrinsic insults, which perturb cellular proteostasis and provoke proteotoxic stress. To counter proteomic perturbations and preserve proteostasis, cells mobilize the proteotoxic stress response (PSR), an evolutionarily conserved transcriptional program mediated by heat shock factor 1 (HSF1). The HSF1-mediated PSR guards the proteome against misfolding and aggregation. In addition to proteotoxic stress, emerging studies reveal that this proteostatic mechanism also responds to cellular energy state. This regulation is mediated by the key cellular metabolic sensor AMP-activated protein kinase (AMPK). In this review, we present an overview of the maintenance of proteostasis by HSF1, the metabolic regulation of the PSR, particularly focusing on AMPK, and their implications in the two major age-related diseases-diabetes mellitus and neurodegenerative disorders.
Collapse
Affiliation(s)
- Kuo-Hui Su
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Chengkai Dai
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA.
| |
Collapse
|