scispace - formally typeset
Search or ask a question

Showing papers in "Cell Stress & Chaperones in 2020"


Journal ArticleDOI
TL;DR: The authors' results show molecular mimicry between the virus and human heat shock proteins 90 and 60, which are associated with Guillain-Barré syndrome and other autoimmune diseases, and embedded in immunoreactive epitopes that have been experimentally validated in the human host.
Abstract: Severe acute respiratory syndrome-related coronavirus 2 infection has been associated with Guillain-Barre syndrome. We investigated here the potential mechanism underlying the virus-induced damage of the peripheral nervous systems by searching the viral amino acid sequence for peptides common to human autoantigens associated with immune-mediated polyneuropathies. Our results show molecular mimicry between the virus and human heat shock proteins 90 and 60, which are associated with Guillain-Barre syndrome and other autoimmune diseases. Crucially, the shared peptides are embedded in immunoreactive epitopes that have been experimentally validated in the human host.

97 citations


Journal ArticleDOI
TL;DR: It is postulate that post-translational modifications, induced by physical (shear) and chemical (metabolic) stress caused respectively by the risk factors hypertension and diabetes, might have a role in determining plasma-cell membrane localization and, in turn, autoimmune-induced endothelial damage.
Abstract: Severe acute respiratory syndrome corona virus 2 (SARS-CoV-2), the cause of COVID-19 disease, has the potential to elicit autoimmunity because mimicry of human molecular chaperones by viral proteins. We compared viral proteins with human molecular chaperones, many of which are heat shock proteins, to determine if they share amino acid-sequence segments with immunogenic-antigenic potential, which can elicit cross-reactive antibodies and effector immune cells with the capacity to damage-destroy human cells by a mechanism of autoimmunity. We identified the chaperones that can putatively participate in molecular mimicry phenomena after SARS-CoV-2 infection, focusing on those for which endothelial cell plasma-cell membrane localization has already been demonstrated. We also postulate that post-translational modifications, induced by physical (shear) and chemical (metabolic) stress caused respectively by the risk factors hypertension and diabetes, might have a role in determining plasma-cell membrane localization and, in turn, autoimmune-induced endothelial damage.

79 citations


Journal ArticleDOI
TL;DR: This paper will discuss how small heat shock proteins give rise to neurodegenerative disorders themselves while it will also highlight how these proteins can fulfil protective functions in neurodegenersative disorders caused by protein aggregation.
Abstract: Small heat shock proteins are ubiquitously expressed chaperones, yet mutations in some of them cause tissue-specific diseases. Here, we will discuss how small heat shock proteins give rise to neurodegenerative disorders themselves while we will also highlight how these proteins can fulfil protective functions in neurodegenerative disorders caused by protein aggregation. The first half of this paper will be focused on how mutations in HSPB1, HSPB3, and HSPB8 are linked to inherited peripheral neuropathies like Charcot-Marie-Tooth (CMT) disease and distal hereditary motor neuropathy (dHMN). The second part of the paper will discuss how small heat shock proteins are linked to neurodegenerative disorders like Alzheimer’s, Parkinson’s, and Huntington’s disease.

51 citations


Journal ArticleDOI
TL;DR: It is proposed that subjects with metabolic syndrome, old age, and male gender have the greatest morbidity and mortality and have low stress proteins, in particular, low intracellular heme oxygenase (HO-1), making them particularly vulnerable to the COVID-19 pandemic.
Abstract: The COVID-19 pandemic needs therapies that are presently available and safe. We propose that subjects with metabolic syndrome, old age, and male gender have the greatest morbidity and mortality and have low stress proteins, in particular, low intracellular heme oxygenase (HO-1), making them particularly vulnerable to the disease. Additionally, COVID-19's heme reduction may contribute to even lower HO-1. Low-grade inflammation associated with these risk factors contributes to triggering a cytokine storm that spreads to multi-organ failure and near death. The high mortality of those treated with ventilator assistance may partially be explained by ventilator-induced inflammation. The cytoprotective and anti-inflammatory properties of HO-1 can limit the infection's damage. A paradox of COVID-19 hospital admissions data suggests that fewer cigarette-smokers are admitted compared with non-smokers in the general population. This unexpected observation may result from smoke induction of HO-1. Therapies with anti-viral properties that raise HO-1 include certain anesthetics (sevoflurane or isoflurane), hemin, estrogen, statins, curcumin, resveratrol, and melatonin. Controlled trials of these HO-1 inducers should be done in order to prevent or treat COVID-19 disease.

49 citations


Journal ArticleDOI
TL;DR: The hypothesis is that the disease escapes from the hands of the doctors who are treating it through a fog of molecular mimicry phenomena: antibodies against the viruses might cross-react with epitopes of self-proteins abnormally expressed on the plasma membrane surface of stressed endothelial cells.
Abstract: It is evident that COVID-19, the disease due to severe acute respiratory syndrome coronavirus 2 (commonly abbreviated SARS-CoV-2) etiological agent, is not uniquely a respiratory disease, at least in a subset of patients. It is possible that we are facing a “proteiform” disease (from Proteus, the sea god who could change shape to avoid capture). As other coronavirusinduced infections, it initially affects the upper airways, but can move abruptly to the lower respiratory tract. Lastly, while physicians treat the bilateral lung pneumonia of their patients, systemic complications can appear characterized by thrombosis, disseminated intravascular coagulation and multi-organ failure. Health systems worldwide are now facing three major problems: 1) to identify asymptomatic carriers of infection; 2) to give effective home care to symptomatic people; 3) to properly stage hospitalized patients to avoid overwhelming numbers in intensive care units. This last group of patients experience, among other clinical signs, very low pO2 blood concentrations due to the destruction of lung parenchyma and massive activation of both innate and acquired immune responses. Presently, only ex juvantibus treatments are done in the most severe cases, since we are far from understanding what primes the systemic vascular complications. The term ex juvantibus or sometimes “ex adiuvantibus” from Latin, meaning “from that which helps” refers, in medical contexts, to the process of making an inference about disease causation from an observed response. I have a modest proposal that I would like to share with our readers. After studying the published clinical reports, what surprised me is that most patients who had the most severe complications were affected by two comorbidities (or their longterm effects): hypertension and diabetes. Both of them induce, among other problems, chronic stress on endothelial cells that in turn can express molecules on their plasma membranes abnormally as an effect of post-translational modifications of intracellular proteins, including some heat shock proteins. This condition can predispose cells and tissues to molecular mimicry phenomena that may occur during an infection (Cappello et al. 2009; Delunardo et al. 2013; Kotlarz et al. 2013; Mayr et al. 1999; Sun et al. 2006). If we add to this chronic stress an acute stress due to the low pO2 blood concentration and systemic inflammation, all the conditions for a perfect storm are present. My hypothesis is that the disease escapes from the hands of the doctors who are treating it through a fog of molecular mimicry phenomena: antibodies against the viruses might cross-react with epitopes of self-proteins abnormally expressed on the plasma membrane surface of stressed endothelial cells. In turn, these autoimmune reactions against endothelium can generate thrombosis, disseminated intravascular coagulation and multi-organ failure. Surprisingly, to date, no items appear on PubMed or other biomedical search engines if we look for “molecular mimicry” and “COVID-19”. Hence, I would like to strongly encourage researchers and physicians to consider this conjecture among others. In vitro and in vivo studies along with bioinformatic analyses can be used to evaluate this hypothesis.

47 citations


Journal ArticleDOI
Chao Jiang1, Ping Luo1, Xian Li1, Ping Liu1, Yong Li, Jie Xu 
TL;DR: Experimental results indicate that curcumin inhibits inflammation, oxidative stress, and the matrix degradation of TMJ inflammatory chondrocytes through the Nrf2/ARE signaling pathway, thereby exerting cartilage protective effects.
Abstract: Temporomandibular joint osteoarthritis (TMJ OA) is a complex multifactorial disease that can be induced by inflammation and oxidative stress. Curcumin has been reported to have anti-inflammatory and antioxidant properties. Herein, the anti-inflammatory and antioxidant mechanisms of curcumin in TMJ OA were investigated. Curcumin treatment inhibited the expression of the inflammation mediators IL-6, iNOS, and COX-2 and of the matrix-degrading proteinases MMP-1, MMP-3, MMP-9, MMP-13, ADAMTS-4, and ADAMTS-5 and upregulated the mRNA levels of the cartilage anabolic factors COL2A1 and ACAN after IL-1β treatment. Curcumin treatment also decreased oxidative stress injury following IL-1β stimulation. Pathway analysis demonstrated that the ROS/Nrf2/HO-1-SOD2-NQO-1-GCLC signaling axis is a key axis through which curcumin activates the Nrf2/ARE pathway in TMJ inflammatory chondrocytes. Curcumin-induced anti-inflammatory and cartilage protective effects were significantly abrogated by specific Nrf2 siRNA. In vivo results demonstrated that curcumin treatment protected TMJ articular cartilage from progressive degradation. Our experimental results indicate that curcumin inhibits inflammation, oxidative stress, and the matrix degradation of TMJ inflammatory chondrocytes through the Nrf2/ARE signaling pathway, thereby exerting cartilage protective effects. This study provides insight into potential therapeutic approaches for TMJ OA.

40 citations


Journal ArticleDOI
TL;DR: Apigenin’s non-toxicity and ability to affect multiple oxidative stress–related parameters through its effects on Nrf2 signaling in melanocytes suggests that it may prove to be a valuable therapeutic tool in long-term management of vitiligo.
Abstract: Vitiligo is a chronic, autoimmune destruction of melanocytes, resulting in progressively expanding depigmented skin patches. Severity of the disorder, which affects approximately 1% of humans, may be mitigated using topical corticosteroids combined with phototherapy; along with other clinical strategies; however, no definitive cures are currently available. Here, the capacity of apigenin, a plant-derived aglycone, to inhibit oxidative stress-mediated melanocyte depletion in vitro using a PIG3V vitiligo perilesional melanocyte cell model is evaluated. PIG3V cells, treated with selected doses of apigenin, were challenged with H2O2, then assessed for viability and the oxidative stress-related parameters: superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GSH-Px), and malondialdehyde (MDA) by enzyme-linked immunoabsorbent assay (ELISA). Additionally, expression of nuclear factor erythroid 2p45 (NF-E2)-related factor 2 (Nrf2) and downstream targets was detected using Western blotting. Outcomes demonstrated that compared with negative control cultures, apigenin-treated cells exhibited enhanced viability. Likewise, apigenin enhanced expression of the cellular anti-oxidants SOD, CAT, and GSH-Px, but inhibited production of MDA, an oxidative stress biomarker. Interestingly, the expression and nuclear localization of the Nrf2 transcription factor, an important regulator oxidative stress and its downstream target genes, was significantly increased by apigenin treatment. Apigenin influence on Nrf2 was further validated by experiments demonstrating that Nrf2 knockdown cells failed to exhibit significant apigenin-mediated effects on cell viability and oxidative stress. Apigenin's non-toxicity and ability to affect multiple oxidative stress-related parameters through its effects on Nrf2 signaling in melanocytes suggests that it may prove to be a valuable therapeutic tool in long-term management of vitiligo.

35 citations


Journal ArticleDOI
TL;DR: It is demonstrated that histone deacetylase (HDAC) inhibitors enable the heat shock response in cultured spinal motor neurons, in a stress-dependent manner, and can improve the efficacy of HSP-inducing drugs in murine spinal cord cultures subjected to thermal or proteotoxic stress.
Abstract: Upregulation of heat shock proteins (HSPs) is an approach to treatment of neurodegenerative disorders with impaired proteostasis. Many neurons, including motor neurons affected in amyotrophic lateral sclerosis (ALS), are relatively resistant to stress-induced upregulation of HSPs. This study demonstrated that histone deacetylase (HDAC) inhibitors enable the heat shock response in cultured spinal motor neurons, in a stress-dependent manner, and can improve the efficacy of HSP-inducing drugs in murine spinal cord cultures subjected to thermal or proteotoxic stress. The effect of particular HDAC inhibitors differed with the stress paradigm. The HDAC6 (class IIb) inhibitor, tubastatin A, acted as a co-inducer of Hsp70 (HSPA1A) expression with heat shock, but not with proteotoxic stress induced by expression of mutant SOD1 linked to familial ALS. Certain HDAC class I inhibitors (the pan inhibitor, SAHA, or the HDAC1/3 inhibitor, RGFP109) were HSP co-inducers comparable to the hydroxyamine arimoclomol in response to proteotoxic stress, but not thermal stress. Regardless, stress-induced Hsp70 expression could be enhanced by combining an HDAC inhibitor with either arimoclomol or with an HSP90 inhibitor that constitutively induced HSPs. HDAC inhibition failed to induce Hsp70 in motor neurons expressing ALS-linked mutant FUS, in which the heat shock response was suppressed; yet SAHA, RGFP109, and arimoclomol did reduce loss of nuclear FUS, a disease hallmark, and HDAC inhibition rescued the DNA repair response in iPSC-derived motor neurons carrying the FUSP525Lmutation, pointing to multiple mechanisms of neuroprotection by both HDAC inhibiting drugs and arimoclomol.

30 citations


Journal ArticleDOI
TL;DR: Leukocyte extravasation has drug targets that should be explored for COVID-19 patients and proteins of the infecting coronavirus interfere with these host responses, possibly causing imbalances that result in immunopathogenesis.
Abstract: The current coronavirus disease 2019 (COVID-19) pandemic has become an unprecedented challenge for the healthcare system worldwide, with an overwhelming number of patients requiring clinical attention and an unacceptably high mortality rate within critical care facilities. COVID-19 is caused by a novel enveloped virus containing a single positive RNA strand, designated severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), first identified in China. The virus shows a high rate of infection, spreading very rapidly among the population that, in this era of swift international connections, has resulted in a global pandemic. The lack of drug treatments and vaccines for this condition has resulted in a mitigation approach to avoid community transmission, consisting of a general lockdown of the populations producing a tremendous burden for the global economy. The first SARS virus, SARS-CoV, also appeared in China and spread to at least 29 countries during 2002–2003. The most frequent cause of death from this virus was respiratory insufficiency with subsequent respiratory failure. What little we know about SARS-CoV-2 is based mainly on SARS-CoV and a small number of other animal coronaviruses. These viruses cause damage to tissues and organs of the infected host by direct infection of target cells or indirectly by prolonged activation of host defense responses. The virus has been detected in the lungs and immune cells of patients who have succumbed to the infection, consistent with direct injury to the pulmonary tissue and activation of the immune response. Activation of the innate immune system stimulates the production of chemokines and cytokines as part of the inflammatory defense response. As a result, neutrophils and macrophages, among other immune system cells, are drawn to infected tissues. Macrophage activation is accompanied by the release of more chemokines creating a two-edged sword capable of both killing virus-infected cells and damaging normal host tissues (reviewed in Perlman and Dandekar 2005). If these activated leukocytes remained at the site of lung infection, the lethality of SARS-CoV-2 virus likely would be lower, but unfortunately, both the chemokine/cytokine “storm” and infected/activated leukocytes released into the circulation by damaged lung tissue affect several organs, of which the brain, kidneys, and heart are particularly susceptible (Huang et al. 2005). Leukocyte extravasation has drug targets that should be explored for COVID-19 patients. As we will discuss in more detail, proteins of the infecting coronavirus interfere with these host responses, possibly causing imbalances that result in immunopathogenesis. The uncertainty about the biology and pathology of a new virus has limited our ability to make predictions about possible treatments and has interfered with the ability of epidemiologists to formulate credible models for the pandemic spread. In contrast, the rapid sequencing of the viral genome has been key in the prompt development of a diagnostic test to detect the presence of the virus. In spite of this information, the prospect of producing an efficient vaccine is still months if not years away. Therefore, immediate solutions are necessary to reduce the burden of this disease. In this regard, an understanding of the basic pathophysiology underlying COVID-19 is critical. Emergency room and critical care doctors and nurses along with hospital staff and EMTs have done heroic work caring for patients and providing essential information on the nature of this chameleon-like illness. Dr. Richard Levitan, an emergency physician at Bellevue Hospital in * Lawrence E. Hightower lawrence.hightower@uconn.edu

30 citations


Journal ArticleDOI
TL;DR: A strong anti-inflammatory effect of this gas which ameliorates various manifestations of inflammation including ROS, NO, TNF-α, and interleukin-6 is demonstrated.
Abstract: In recent years, the gasotransmitter hydrogen sulfide (H2S) has been recognized as a biological mediator of immense importance both in eukaryotes and prokaryotes (Kimura 2014; Xiao et al. 2018). H2S is produced in the cells mostly through the reverse transsulfuration pathway (TSP). Transsulfuration is a vital metabolic process common in prokaryotes and eukaryotes that have been studied in detail in mammals including human and several other organisms. It has been demonstrated by different groups that defects in the H2S synthesizing enzyme system are involved in a plethora of diseases in humans including cancer and a number of neurodegenerative diseases (Wallace and Wang 2015; Bhattacharyya et al. 2016). Although at high concentration H2S is a poison, at low concentrations, it elicits cytoprotection during oxidative stress by decreasing reactive oxygen species (ROS) production in a wide range of physiologic and pathologic conditions (Kaya-Yasar et al. 2017; Faller et al. 2018). It is interesting that cysteine is sulfur-rich and likely involved as a modulator of ROS due to S–S bonds. To this end, sulfide-rich water in baths is routinely used in sanatoriums to treat multiple diseases. In recent decades, we studied the effects of endogenous and exogenous hydrogen sulfide at the cellular and organism levels (Yurinskaya et al. 2020; Shilova et al. 2020; Zatsepina et al. in press). In our investigation, we explored slowand fast-releasing H2S donors as well as deletions of the genes responsible for H2S production and demonstrated a strong anti-inflammatory effect of this gas which ameliorates various manifestations of inflammation including ROS, NO, TNF-α, and interleukin-6. Along these lines, there are studies demonstrating antiviral and anti-inflammatory activity of H2S in several rodent models (Bazhanov et al. 2018; Bazhanov et al. 2017). In an animal model, hydrogen sulfide donors are usually introduced by inhalation to efficiently alleviate lung injury and pneumonia induced by bacteria or viruses (Zhang et al. 2019; Sakaguchi et al. 2014; Kakinohana et al. 2019). It was also shown in rodent models that preand posttreatment with hydrogen sulfide prevents ventilator-induced lung injury by limiting inflammation and oxidation (Faller et al. 2017). Since hydrogen sulfide is a toxic gas, its use “as is” for inhalation is problematic. Water-soluble sulfide salts such as Na2S and NaHS generate free H2S in aqueous solutions. Thus, diluted solutions of inorganic sulfides could be used for inhalation with a nebulizer, but an important drawback of these compounds is their regulatory status. There are no clinical data nor any documented evidence of sulfide inhalation, and it is important to note that pharmaceutical grade sulfides are not available. For these reasons, we focused on sodium thiosulfate (Na2S2O3) that is an FDAapproved drug used in the treatment of cyanide poisoning, certain extravasation injuries, and for calciphylaxis associated with chronic kidney disease. USP grade sodium thiosulfate (STS) is available as a drug substance and also as a sterile solution for injections. The molecule of Na2S2O3 contains divalent sulfur (S ) and could be a slow donor of hydrogen sulfide in aqueous solutions and biological systems. This compound is also a potent reducing agent and scavenger of ROS reducing Fe+3 to Fe+2. Sodium thiosulfate is a harmless substance which has been approved by the FDA and used for decades for the treatment of cyanide poisoning (Bebarta et al. 2017). Importantly, sodium thiosulfate was widely used in model organisms to mitigate lung injury (Zhang et al. 2019). Thiosulfate can produce H2S through a nonenzymatic or by an enzymatic pathway (Snijder et al. 2015; Leskova et al. 2017) and, hence, may be successfully applied in humans not only by inhalation but orally and intravenously as well (Farese et al. 2011). In * Mikhail B. Evgen’ev misha672011@yahoo.com

29 citations


Journal ArticleDOI
TL;DR: It is demonstrated that depletion of Nrf2 and GSH was associated with the elevation of oxidative stress and the activation of intrinsic apoptosis in mouse kidney treated with DEHP.
Abstract: Environmental toxicants such as phthalate have been involved in multiple health disorders including renal diseases. Oxidative damage is implicated in many alterations caused by phthalate especially the di(2-ethylhexyl) phthalate (DEHP), which is the most useful phthalate. However, information regarding its mechanism of renal damage is lacking. The transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) regulates gene expression implicated in free radical scavenging and cytoprotection including the antioxidant glutathione (GSH) pathway. The aim of this study was to assess whether DEHP affects the Nrf2 pathway and the GSH concentration. Mice were divided into four groups: a control group and three groups treated with DEHP at different concentrations (5, 50, and 200 mg/kg body weight) for 30 days. Our results showed that DEHP altered the normal levels of serum biochemical parameters creatinine (CREA), urea, and lactate dehydrogenase (LDH). This phthalate caused oxidative damage through the induction of lipid peroxidation and protein oxidation as marked by increase of protein carbonyl (PC) and loss of protein-bound sulfhydryls (PSH). Simultaneously, DEHP treatment decreased the protein level of Nrf-2, HO-1, and GCLC (responsible of GSH synthesis) and decreased the GSH level. Inhibition of the Nrf2 pathway is related to the activation of the mitochondrial pathway of apoptosis. This apoptotic process is evidenced by an upregulation of p53 and Bax protein levels in addition to a downregulation of Bcl-2. Collectively, our data demonstrated that depletion of Nrf2 and GSH was associated with the elevation of oxidative stress and the activation of intrinsic apoptosis in mouse kidney treated with DEHP.

Journal ArticleDOI
TL;DR: The anti-cancer effects of those compounds appear to be associated with anti-inflammatory properties as mentioned in this paper, thus, ongoing laborious efforts investigate the possible application of those agents towards inflammatory disorders of the lungs, such as acute respiratory distress syndrome.
Abstract: Heat shock protein 90 is a highly conserved molecular chaperone, essential for cellular survival under diverse environments. Since this protein is employed by tumors to promote their prevalence, heat shock protein 90 inhibitors have been developed to oppose malignancies. The anti-cancer effects of those compounds appear to be associated with anti-inflammatory properties. Thus, ongoing laborious efforts investigate the possible application of those agents towards inflammatory disorders of the lungs, such as the acute respiratory distress syndrome.

Journal ArticleDOI
TL;DR: This review explores how the structure of αB-c confers its function and interactions within its oligomeric self, with other sHsps, and with aggregation-prone target proteins and highlights the remarkably diverse capabilities of β-c as a caretaker of the cell.
Abstract: In vivo, small heat-shock proteins (sHsps) are key players in maintaining a healthy proteome. αB-crystallin (αB-c) or HspB5 is one of the most widespread and populous of the ten human sHsps. Intracellularly, αB-c acts via its molecular chaperone action as the first line of defence in preventing target protein unfolding and aggregation under conditions of cellular stress. In this review, we explore how the structure of αB-c confers its function and interactions within its oligomeric self, with other sHsps, and with aggregation-prone target proteins. Firstly, the interaction between the two highly conserved regions of αB-c, the central α-crystallin domain and the C-terminal IXI motif and how this regulates αB-c chaperone activity are explored. Secondly, subunit exchange is rationalised as an integral structural and functional feature of αB-c. Thirdly, it is argued that monomeric αB-c may be its most chaperone-species active, but at the cost of increased hydrophobicity and instability. Fourthly, the reasons why hetero-oligomerisation of αB-c with other sHsps is important in regulating cellular proteostasis are examined. Finally, the interaction of αB-c with aggregation-prone, partially folded target proteins is discussed. Overall, this paper highlights the remarkably diverse capabilities of αB-c as a caretaker of the cell.

Journal ArticleDOI
TL;DR: An overview of the available knowledge on the structural complexity of human small heat shock proteins is given to aid the search for therapeutic molecules that could affect the chaperone activity or compensate specific mutations.
Abstract: Small heat shock proteins function as chaperones by binding unfolding substrate proteins in an ATP-independent manner to keep them in a folding-competent state and to prevent irreversible aggregation. They play crucial roles in diseases that are characterized by protein aggregation, such as neurodegenerative and neuromuscular diseases, but are also involved in cataract, cancer, and congenital disorders. For this reason, these proteins are interesting therapeutic targets for finding molecules that could affect the chaperone activity or compensate specific mutations. This review will give an overview of the available knowledge on the structural complexity of human small heat shock proteins, which may aid in the search for such therapeutic molecules.

Journal ArticleDOI
Guo-Dan Liu1, Shi-Feng Zhou1, Xinge Li1, Xuchen Ding1, Miao Tian1 
TL;DR: circ_0002570 might serve as a potential target for diagnosis and treatment for DR by suppressing the abilities of proliferation, migration, and angiogenesis in hRMECs induced by high glucose, which was dependent on miR-1243-angiomotin axis.
Abstract: Diabetic retinopathy (DR) is the most severe microvascular complication of diabetes and a major cause of visual impairment and blindness. However, the treatment for DR is still limited. Our study aimed to explore the role of circular RNA_0002570 in DR. First, we predicted the potential microRNA and mRNA that could bind to circ_0002570 and identified the miR-1243 and angiomotin gene; then, we used RT-PCR and Western blot to measure their expression. Next, we evaluated the abilities of proliferation, migration, and angiogenesis in vitro in human retinal microvascular endothelial cells (hRMECs) by CCK-8, transwell assay, and tube formation assay, respectively. To analyze the relationship among miR-1243, circ_0002570, and angiomotin, RNA pull-down and luciferase assay were performed. Our results showed that, in DR patients and high-glucose–induced hRMECs, miR-1243, circ_0002570, and angiomotin were all abnormally expressed. MiR-1243 could directly and competitively bind to both circ_0002570 and angiomotin mRNA to inhibit their expression. Moreover, circ_0002570 suppressed the abilities of proliferation, migration, and angiogenesis in hRMECs induced by high glucose, which was dependent on miR-1243-angiomotin axis. Furthermore, circ_0002570 could upregulate angiomotin by targeting miR-1243 to mediate the dysfunction of hRMECs induced by high glucose. In conclusion, circ_0002570 might serve as a potential target for diagnosis and treatment for DR.

Journal ArticleDOI
TL;DR: It is suggested that small heat-shock proteins interact with mechanosensitive proteins to regulate physiological extension and contraction cycles and expand the understanding of the activity and functions of sHSPs, and of the roles played by chaperones in general.
Abstract: The ability of cells to respond to stress is central to health. Stress can damage folded proteins, which are vulnerable to even minor changes in cellular conditions. To maintain proteostasis, cells have developed an intricate network in which molecular chaperones are key players. The small heat-shock proteins (sHSPs) are a widespread family of molecular chaperones, and some sHSPs are prominent in muscle, where cells and proteins must withstand high levels of applied force. sHSPs have long been thought to act as general interceptors of protein aggregation. However, evidence is accumulating that points to a more specific role for sHSPs in protecting proteins from mechanical stress. Here, we briefly introduce the sHSPs and outline the evidence for their role in responses to mechanical stress. We suggest that sHSPs interact with mechanosensitive proteins to regulate physiological extension and contraction cycles. It is likely that further study of these interactions - enabled by the development of experimental methodologies that allow protein contacts to be studied under the application of mechanical force - will expand our understanding of the activity and functions of sHSPs, and of the roles played by chaperones in general.

Journal ArticleDOI
TL;DR: The results suggest hypoxia leads to oxidative damage and compensation mechanism of the brain, heart, lung, liver, and kidney in varying degrees of mice.
Abstract: This study was first and systematically conducted to evaluate the hypoxia response of the brain, heart, lung, liver, and kidney of mice exposed to an animal hypobaric chamber. First, we examined the pathological damage of the above tissues by Hematoxylin & eosin (H&E) staining. Secondly, biochemical assays were used to detect oxidative stress indicators such as superoxide dismutase (SOD), malondialdehyde (MDA), reduced glutathione (GSH), and oxidized glutathione (GSSG). Finally, the hypoxia compensation mechanism of tissues was evaluated by expression levels of hypoxia-inducible factor 1 alpha (HIF-1α), erythropoietin (EPO), and vascular endothelial growth factor (VEGF). During the experiment, the mice lost weight gradually on the first 3 days, and then, the weight loss tended to remain stable, and feed consumption showed the inverse trend. H&E staining results showed that there were sparse and atrophic neurons and dissolved chromatin in the hypoxia group. And hyperemia occurred in the myocardium, lung, liver, and kidney. Meanwhile, hypoxia stimulated the enlargement of myocardial space, the infiltration of inflammatory cells in lung tissue, the swelling of epithelial cells in hepatic lobules and renal tubules, and the separation of basal cells. Moreover, hypoxia markedly inhibited the activity of SOD and GSH and exacerbated the levels of MDA and GSSG in the serum and five organs. In addition, hypoxia induced the expression of HIF-1α, EPO, and VEGF in five organs. These results suggest hypoxia leads to oxidative damage and compensation mechanism of the brain, heart, lung, liver, and kidney in varying degrees of mice.

Journal ArticleDOI
TL;DR: HSP70 played critical roles in maintaining the cellular homeostasis of plants during adaptation to drought and under normal plant life conditions and it was speculated that proline was, to some extent, involved in improving the loss of protein folding or function resulting from HSP70 deficiency.
Abstract: The 70-kDa heat shock proteins (HSP70s) are a conserved class of chaperones that play critical roles during the normal life cycle of plants. HSP70s are particularly involved in the regulation of biotic and abiotic stress responses. In this paper, the potential roles of this protein were investigated. A reverse genetic approach was employed for transient silencing of hsp70 gene in tomato (Solanum lycopersicum L.) to evaluate different growth and physiological parameters under normal conditions and during the response to drought stress. A combined ANOVA (analysis of variance) and HCA (hierarchical clustering analysis) showed that hsp70 silencing led to severe growth retardation and mortality, significant membrane damage and leakage, decline in relative water content, low rate of pigment accumulation, and reduced antioxidant enzyme activity under normal and drought stress conditions. Among the different parameters, proline was the only trait that was unaffected by gene silencing and accumulated by similar amounts to that of nonsilent plants. In conclusion, HSP70 played critical roles in maintaining the cellular homeostasis of plants during adaptation to drought and under normal plant life conditions. It was speculated that proline was, to some extent, involved in improving the loss of protein folding or function resulting from HSP70 deficiency, and played a crucial role in the adaptation of plants on exposure to stress.

Journal ArticleDOI
TL;DR: The disorders with abnormal FABPs as chaperonopathies in a distinct nosological entity are assembled within of a coherent nosological group, focusing on abnormal lipid chaperones as biomarkers of disease and etiological-pathogenic factors.
Abstract: Fatty acid-binding proteins (FABPs) are lipid chaperones assisting in the trafficking of long-chain fatty acids with functions in various cell compartments, including oxidation, signaling, gene-transcription regulation, and storage. The various known FABP isoforms display distinctive tissue distribution, but some are active in more than one tissue. Quantitative and/or qualitative changes of FABPs are associated with pathological conditions. Increased circulating levels of FABPs are biomarkers of disorders such as obesity, insulin resistance, cardiovascular disease, and cancer. Deregulated expression and malfunction of FABPs can result from genetic alterations or posttranslational modifications and can be pathogenic. We have assembled the disorders with abnormal FABPs as chaperonopathies in a distinct nosological entity. This entity is similar but separate from that encompassing the chaperonopathies pertaining to protein chaperones. In this review, we discuss the role of FABPs in the pathogenesis of metabolic syndrome, cancer, and neurological diseases. We highlight the opportunities for improving diagnosis and treatment that open by encompassing all these pathological conditions within of a coherent nosological group, focusing on abnormal lipid chaperones as biomarkers of disease and etiological-pathogenic factors.

Journal ArticleDOI
TL;DR: It is concluded that HsfA1d plays a vital role in plant thermotolerance and hence can be effectively used to enhance the resistance of crop plants against heat stress.
Abstract: Thermal stress induces a wide array of morphological and physiological changes in potato affecting its development and economic yield. Response to thermal stress in plants is mostly regulated by heat shock factors (hsfs). The current study aimed at improving heat tolerance by transforming potato plant with heat shock factor, HsfA1d, using Agrobacterium. Gateway cloning strategy was adopted for isolation of HsfA1d from Arabidopsis thaliana and cloning into plant expression vector. The target gene was introduced into potato by infecting internodal explants with Agrobacterium strain GV3101 carrying pGWB402Ω-HsfA1d construct. Upon exposure to heat stress, the wild-type plants turned yellowish, whereas no phenotypic effect on transgenic plants was observed. Expression of HsfA1d in transgenic plants was increased by 5.8-fold under thermal stress compared to room temperature. Transgenic plants exhibited 6-fold increase in the expression of downstream HSP70 under thermal stress compared to wild-type plants. Both chlorophyll a and b were significantly decreased in wild-type plants while no such decrease was recorded in transgenic plants under thermal stress. Heat stress was found to have no significant effect on carotenoid pigments of both wild-type and transgenic plants. Significantly lower electrolyte leakage from transgenic plants was witnessed compared to wild type upon exposure to thermal stress. Transgenic plants accumulated significantly higher proline content compared to wild-type plants under heat stress. It is concluded that HsfA1d plays a vital role in plant thermotolerance and hence can be effectively used to enhance the resistance of crop plants against heat stress.

Journal ArticleDOI
TL;DR: A unique anti-tumor mechanism of MAN is revealed by provoking ROS production through downregulation of NAMPT/NAD signaling and further validated MAN as a potential therapeutic reagent for lung cancer treatment.
Abstract: α-Mangostin (MAN) is a bioactive compound isolated from the inedible pericarp of a tropical fruit mangosteen (Garcinia mangostana Linn). It exhibits notable therapeutic potentials on lung cancers, but the underlying mechanisms are still largely unknown. This study was designed to further explore the mechanisms involved in cytotoxicity of MAN on A549 cells. Apoptosis and cell cycle distribution were analyzed by flow cytometry methods. The fluorescent probes DCFH-DA and JC-1 were used to assess the intracellular reactive oxidative species (ROS) and mitochondrial membrane potential statuses, respectively. The regulation of MAN on relevant pathways was investigated by immunoblotting assays. The results obtained indicated that MAN caused significant apoptosis and cell cycle arrest in A549 cells, which eventually resulted in inhibition on cell proliferation in vitro. All these phenomena were synchronized with escalated oxidative stress and downregulation of nicotinamide phosphoribosyltransferase/nicotinamide adenine dinucleotide (NAMPT/NAD). Supplementation with nicotinamide mononucleotide (NMN) and N-acetylcysteine (NAC) efficiently eased MAN-induced ROS accumulation, and potently antagonized MAN-elicited apoptosis and cell cycle arrest. The pro-apoptotic effect of MAN was further confirmed by increased expressions of cleaved caspase 3, 6, 7, and 9, and its effect on cell cycle progression was validated by the altered expressions of p-p38, p-p53, CDK4, and cyclin D1. The immunoblotting assays also demonstrated that NAC/NMN effectively restored these molecular changes elicited by MAN treatment. Collectively, this study revealed a unique anti-tumor mechanism of MAN by provoking ROS production through downregulation of NAMPT/NAD signaling and further validated MAN as a potential therapeutic reagent for lung cancer treatment.

Journal ArticleDOI
TL;DR: The proteome and protein-protein interaction network of the Himalayan psychrophilic P. helmanticensis under cold stress is Documented, revealing that Tif, Tig, DnaK, and Adk were crucial proteins involved in cold adaptation.
Abstract: Himalayan mountains are distinctly characterized for their unique climatic and topographic variations; therefore, unraveling the cold-adaptive mechanisms and processes of native life forms is always being a matter of concern for scientific community. In this perspective, the proteomic response of psychrophilic diazotroph Pseudomonas helmanticensis was studied towards low-temperature conditions. LC-MS-based analysis revealed that most of the differentially expressed proteins providing cold stress resistance were molecular chaperons and cold shock proteins. Enzymes involved in proline, polyamines, unsaturated fatty acid biosynthesis, ROS-neutralizing pathways, and arginine degradation were upregulated. However, proteins involved in the oxidative pathways of energy generation were severalfold downregulated. Besides these, the upregulation of uncharacterized proteins at low temperature suggests the expression of novel proteins by P. helmanticensis for cold adaptation. Protein interaction network of P. helmanticensis under cold revealed that Tif, Tig, DnaK, and Adk were crucial proteins involved in cold adaptation. Conclusively, this study documents the proteome and protein-protein interaction network of the Himalayan psychrophilic P. helmanticensis under cold stress.

Journal ArticleDOI
TL;DR: Investigation of the renoprotective effect of H 2 S, in a renal injury model, and its crosstalk with other gasotransmitters such as CO found improvement in kidney function produced by NaHS was reduced by combination with ZnPP but still improved as compared with the GEN-treated group.
Abstract: Hydrogen sulfide (H2S), along with nitric oxide (NO) and carbon monoxide (CO), proved to have renoprotective effects in various renal diseases. Therefore, this study investigated the renoprotective effect of H2S, in a renal injury model, and its crosstalk with other gasotransmitters such as CO. Thirty-two adult rats were divided into four groups: control, gentamicin (GEN)-treated, GEN + sodium hydrosulfide (NaHS), and GEN + NaHS + zinc protoporphyrin (ZnPP) groups. GEN was used to induce renal injury, NaHS is a water-soluble H2S, and ZnPP is a selective heme oxygenase-1 (HO-1) inhibitor used to inhibit CO synthesis in vivo. NaHS improved kidney functions in the GEN group as evidenced by significantly lower levels of renal injury markers: serum urea, creatinine, uric acid, urinary albumin excretion, and urinary albumin/creatinine. Moreover, NaHS administration to the GEN-treated group significantly lowered renal levels of NO and tumor necrosis factor-α with an increase in total antioxidant, HO-1, and interleukin-10 levels. Furthermore, NaHS administration downregulated the GEN-induced overexpression of the renal inducible nitric oxide synthase (iNOS) and upregulated the suppression of endothelial nitric oxide synthase (eNOS) with improvement in the histological examination and periodic acid Schiff (PAS) staining. However, this improvement in kidney function produced by NaHS was reduced by combination with ZnPP but still improved as compared with the GEN-treated group. The renoprotective effects of H2S can be through its effects on renal tissue antioxidants, pro-inflammatory and anti-inflammatory cytokines, and expression of eNOS and iNOS which can be partially dependent on CO pathway via induction of HO-1 enzyme.

Journal ArticleDOI
TL;DR: Golden and brown scallops were subjected to acute thermal challenge, and the LTE50 and LTI50 as well as the correlation between CnHSP90 gene expression and TCC were determined, suggesting that both carotenoids and HSP90 levels could improve thermal resistance in the noble scallop.
Abstract: The noble scallop Chlamys nobilis is an economically important marine bivalve cultivated in the southern sea of China since the 1980s. Unfortunately, mass mortality of this scallop species often occurs in summer. The present study was conducted to investigate whether the expression of heat shock protein 90 (HSP90) and level of carotenoids could enhance high-temperature stress resistance in scallop. First, the HSP90 homolog of C. nobilis (designated CnHSP90) was identified and cloned. The complete cDNA sequence of CnHSP90 was 2631 bp, including a 2181-bp open reading frame (ORF) encoding a 726 amino acid polypeptide with five HSP90 family signatures, and sharing high homology with members of the HSP90 family. CnHSP90 was ubiquitously expressed in all examined tissues including the intestine, kidney, adductor, mantle, gill, and gonad, with the highest in the gonad. Golden and brown scallops, which contain significantly different total carotenoid content (TCC), were subjected to acute thermal challenge, and the LTE50 (semi-lethal temperature at 36 h heat shock) and LTI50 (semi-lethal time after heat shock) as well as the correlation between CnHSP90 gene expression and TCC were determined. The LTE50 of golden scallop (32.14 °C) was higher than that of brown scallops (31.19 °C), with longer LTI50 at all tested temperatures, indicating that golden scallops were more resistant to thermal stress than brown scallops. Similarly, the mRNA expression levels of CnHSP90 in gill of golden scallops were significantly higher (P < 0.05) than that of brown scallops at 6, 12, 24, and 36 h, with a strong positive correlation between CnHSP90 expression level and TCC. This suggests that both carotenoids and HSP90 levels could improve thermal resistance in the noble scallops.

Journal ArticleDOI
Abdo A. Elfiky1
TL;DR: Results show a promising binding site that supports the hypothesis of HSPA5 selectivity for binding to a specific peptide sequence (pep42) and paves the way to suggest possible inhibitors to stop viral association with cell-surface receptors and subsequently reduce viral infection.
Abstract: Ebola virus (EBOV) infection is a widespread infection that has created a bad memory in Africa. In the 2014 and 2015 outbreak, more than 28,000 infections were reported by the World Health Organization, with about 11,300 deaths in Guinea, Liberia, and Sierra Leone. Heat shock protein A5 (HSPA5), termed also GRP78, is a host cell chaperone protein responsible for the unfolded protein response in the endoplasmic reticulum. Under stress, HSPA5 is upregulated and becomes cell-surface exposed. Recent studies report the association of cell-surface HSPA5 with EBOV glycoproteins GP1 and GP2. In this study, structural and sequence analysis and molecular docking are used to predict the possible binding site between the cell-surface HSPA5 and EBOV GP1. The results show a promising binding site that supports the hypothesis of HSPA5 selectivity for binding to a specific peptide sequence (pep42). This study paves the way to suggest possible inhibitors to stop viral association with cell-surface receptors and subsequently reduce viral infection.

Journal ArticleDOI
TL;DR: The roles of PP5 in cancer progression and survival are reviewed and the unique features of the PP5 structure that differentiate it from other phosphoprotein phosphatase (PPP) family members and make it an attractive therapeutic target are discussed.
Abstract: Protein phosphatase 5 (PP5) is a serine/threonine protein phosphatase that regulates many cellular functions including steroid hormone signaling, stress response, proliferation, apoptosis, and DNA repair. PP5 is also a co-chaperone of the heat shock protein 90 molecular chaperone machinery that assists in regulation of cellular signaling pathways essential for cell survival and growth. PP5 plays a significant role in survival and propagation of multiple cancers, which makes it a promising target for cancer therapy. Though there are several naturally occurring PP5 inhibitors, none is specific for PP5. Here, we review the roles of PP5 in cancer progression and survival and discuss the unique features of the PP5 structure that differentiate it from other phosphoprotein phosphatase (PPP) family members and make it an attractive therapeutic target.

Journal ArticleDOI
TL;DR: It is found that HSP70 inhibitor VER-155008 alone and the combination of bortezomib synergistically induced MM cell apoptosis markedly in vitro, and this finding may support for combination therapy in MM patients in the future.
Abstract: Proteasome inhibitor bortezomib is one of the most effective drugs currently available for the treatment of multiple myeloma (MM). However, the intrinsic and acquired resistance to bortezomib can limit its effectiveness. The activation of heat shock response has been characterized as a potential resistance mechanism protecting MM cells from bortezomib-induced cell death. In this study, in response to bortezomib therapy, we discovered that HSP70 is one of the most substantially upregulated heat shock proteins. In order to further explore approaches to sensitizing bortezomib-based treatment for MM, we investigated whether targeting HSP70 using a specific inhibitor VER-155008 combined with bortezomib could overcome the acquired resistance in MM. We found that HSP70 inhibitor VER-155008 alone significantly decreased MM cell viability. Moreover, the combination of VER-155008 and bortezomib synergistically induced MM cell apoptosis markedly in vitro. Notably, the combined treatment was found to increase the cleavage of PARP, an early marker of chemotherapy-induced apoptosis. Importantly, the reduction of anti-apoptotic Bcl-2 family member Bcl-2, Bcl-xL, and Mcl-1 and the induction of pro-apoptotic Bcl-2 family member BH3-only protein NOXA and Bim were confirmed to be tightly associated with the synergism. Finally, the ER stress marker CHOP (CCAAT-enhancer binding protein homologous protein), which can cause transcriptional activation of genes involved in cell apoptosis, was markedly induced by both VER-155008 and bortezomib. Taken together, our finding of a strong synergistic interaction between VER-155008 and bortezomib may support for combination therapy in MM patients in the future.

Journal ArticleDOI
TL;DR: While the mechanisms of pathological mutations in HspB1 are likely to vary greatly across different mutations, further in vitro and in vivo studies are required for a better understanding of the CMT disease at molecular level.
Abstract: Charcot-Marie-Tooth (CMT) disease is major hereditary neuropathy. CMT has been linked to mutations in a range of proteins, including the small heat shock protein HspB1. Here we review the properties of several HspB1 mutants associated with CMT. In vitro, mutations in the N-terminal domain lead to a formation of larger HspB1 oligomers when compared with the wild-type (WT) protein. These mutants are resistant to phosphorylation-induced dissociation and reveal lower chaperone-like activity than the WT on a range of model substrates. Mutations in the α-crystallin domain lead to the formation of yet larger HspB1 oligomers tending to dissociate at low protein concentration and having variable chaperone-like activity. Mutations in the conservative IPV motif within the C-terminal domain induce the formation of very large oligomers with low chaperone-like activity. Most mutants interact with a partner small heat shock protein, HspB6, in a manner different from that of the WT protein. The link between the altered physico-chemical properties and the pathological CMT phenotype is a subject of discussion. Certain HspB1 mutations appear to have an effect on cytoskeletal elements such as intermediate filaments and/or microtubules, and by this means damage the axonal transport. In addition, mutations of HspB1 can affect the metabolism in astroglia and indirectly modulate the viability of motor neurons. While the mechanisms of pathological mutations in HspB1 are likely to vary greatly across different mutations, further in vitro and in vivo studies are required for a better understanding of the CMT disease at molecular level.

Journal ArticleDOI
TL;DR: There was a significant effect of heat acclimation on HSP70 induction in humans and the only factor among identified studies that may moderate this response was the frequency of days of heat exposure.
Abstract: Heat acclimation (HA) in humans promotes thermoregulatory adaptations that support management of core temperature in hot environments and reduces the likelihood of heat related illness. Another adaptation to HA is thermotolerance through induction of the heat shock protein (HSP) stress system, which provides protection against thermal insult. However, whether or not HA leads to upregulation of the intracellular HSP system, namely intracellular HSP70 (HSP70), is unclear in humans. Therefore, the purposes of this meta-analysis were to determine if HA leads to HSP70 induction among humans and to evaluate how methodological differences among HA studies influence findings regarding HA-induced HSP70 accumulation. Several databases were searched to identify studies that measured HSP70 (protein and mRNA) changes in response to HA among humans. The effect of HA on HSP70 was analyzed. Differences in the effect of HA were assessed between protein and mRNA. The moderating effect of several independent variables (HA frequency, HA duration, core temperature, exercise intensity) on HSP70 was also evaluated. Data were extracted from 12 studies including 118 participants (mean age 24 years, 98% male). There was a significant effect of HA on HSP70 expression, g = 0.97 (95% CI, 0.08–1.89). The effect of HA was different between subgroups (protein vs. mRNA), g = 1.51 (95% CI, 0.71–2.31), and g = − 0.39 (95% CI, − 1.36), respectively. The frequency of HA (in days) moderated HSP70 protein expression. There was a significant effect of heat acclimation on HSP70 induction in humans. The only factor among identified studies that may moderate this response was the frequency (number of days) of heat exposure.

Journal ArticleDOI
TL;DR: Interestingly, it is found that the interaction of HSPA5 with negatively charged liposomes promotes an oligomerization process via intermolecular disulfide bonds in which the N-terminus end of the protein plays a critical role.
Abstract: Heat shock proteins (HSPs) are ubiquitous polypeptides expressed in all living organisms that participate in several basic cellular processes, including protein folding, from which their denomination as molecular chaperones originated. There are several HSPs, including HSPA5, also known as 78-kDa glucose-regulated protein (GRP78) or binding immunoglobulin protein (BIP) that is an ER resident involved in the folding of polypeptides during their translocation into this compartment prior to the transition to the Golgi network. HSPA5 is detected on the surface of cells or secreted into the extracellular environment. Surface HSPA5 has been proposed to have various roles, such as receptor-mediated signal transduction, a co-receptor for soluble ligands, as well as a participant in tumor survival, proliferation, and resistance. Recently, surface HSPA5 has been reported to be a potential receptor of some viruses, including the novel SARS-CoV-2. In spite of these observations, the association of HSPA5 within the plasma membrane is still unclear. To gain information about this process, we studied the interaction of HSPA5 with liposomes made of different phospholipids. We found that HSPA5 has a high affinity for negatively charged phospholipids, such as palmitoyl-oleoyl phosphoserine (POPS) and cardiolipin (CL). The N-terminal and C-terminal domains of HSPA5 were independently capable of interacting with negatively charged phospholipids, but to a lesser extent than the full-length protein, suggesting that both domains are required for the maximum insertion into membranes. Interestingly, we found that the interaction of HSPA5 with negatively charged liposomes promotes an oligomerization process via intermolecular disulfide bonds in which the N-terminus end of the protein plays a critical role.