scispace - formally typeset
Search or ask a question

Showing papers in "Biofactors in 2021"


Journal ArticleDOI
TL;DR: Luteolin can improve cognitive decline and enhance neuroprotection in neurodegenerative diseases, TBI, and stroke, and recent reports suggest that luteolin suppresses systemic and neuroinflammatory responses in Coronavirus disease 2019.
Abstract: Neuroinflammation leads to neurodegeneration, cognitive defects, and neurodegenerative disorders. Neurotrauma/traumatic brain injury (TBI) can cause activation of glial cells, neurons, and neuroimmune cells in the brain to release neuroinflammatory mediators. Neurotrauma leads to immediate primary brain damage (direct damage), neuroinflammatory responses, neuroinflammation, and late secondary brain damage (indirect) through neuroinflammatory mechanism. Secondary brain damage leads to chronic inflammation and the onset and progression of neurodegenerative diseases. Currently, there are no effective and specific therapeutic options to treat these brain damages or neurodegenerative diseases. Flavone luteolin is an important natural polyphenol present in several plants that show anti-inflammatory, antioxidant, anticancer, cytoprotective, and macrophage polarization effects. In this short review article, we have reviewed the neuroprotective effects of luteolin in neurotrauma and neurodegenerative disorders and pathways involved in this mechanism. We have collected data for this study from publications in the PubMed using the keywords luteolin and mast cells, neuroinflammation, neurodegenerative diseases, and TBI. Recent reports suggest that luteolin suppresses systemic and neuroinflammatory responses in Coronavirus disease 2019 (COVID-19). Studies have shown that luteolin exhibits neuroprotective effects through various mechanisms, including suppressing immune cell activation, such as mast cells, and inflammatory mediators released from these cells. In addition, luteolin can suppress neuroinflammatory response, activation of microglia and astrocytes, oxidative stress, neuroinflammation, and the severity of neuroinflammatory diseases such as Alzheimer's disease, Parkinson's disease, multiple sclerosis, and TBI pathogenesis. In conclusion, luteolin can improve cognitive decline and enhance neuroprotection in neurodegenerative diseases, TBI, and stroke.

82 citations


Journal ArticleDOI
TL;DR: In this article, a phytosomal formulation (in olive pomace oil) of the natural flavonoid luteolin was used to mitigate brain fog in patients undergoing or following chemotherapy for cancer (chemofog or chemobrain), as well in patients with myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) or mast cell activation syndrome (MCAS).
Abstract: COVID-19 leads to severe respiratory problems, but also to long-COVID syndrome associated primarily with cognitive dysfunction and fatigue. Long-COVID syndrome symptoms, especially brain fog, are similar to those experienced by patients undertaking or following chemotherapy for cancer (chemofog or chemobrain), as well in patients with myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) or mast cell activation syndrome (MCAS). The pathogenesis of brain fog in these illnesses is presently unknown but may involve neuroinflammation via mast cells stimulated by pathogenic and stress stimuli to release mediators that activate microglia and lead to inflammation in the hypothalamus. These processes could be mitigated by phytosomal formulation (in olive pomace oil) of the natural flavonoid luteolin.

81 citations


Journal ArticleDOI
TL;DR: Luteolin belongs to the group of flavonoids and can be found in flowers, herbs, vegetables and spices as discussed by the authors, which plays an important role in defending plants, for example against UV radiation by partially absorbing UVA and UVB radiation.
Abstract: Luteolin belongs to the group of flavonoids and can be found in flowers, herbs, vegetables and spices. It plays an important role in defending plants, for example against UV radiation by partially absorbing UVA and UVB radiation. Thus, luteolin can also decrease adverse photobiological effects in the skin by acting as a first line of defense. Furthermore, anti-oxidative and anti-inflammatory activities of luteolin were described on keratinocytes and fibroblasts as well as on several immune cells (e.g., macrophages, mast cell, neutrophils, dendritic cells and T cells). Luteolin can suppress proinflammatory mediators (e.g., IL-1β, IL-6, IL-8, IL-17, IL-22, TNF-α and COX-2) and regulate various signaling pathway (e.g., the NF-κB, JAK-STAT as well as TLR signaling pathway). In this way, luteolin modulates many inflammatory processes of the skin. The present review summarizes the recent in vitro and in vivo research on luteolin in the field of skin aging and skin cancer, wound healing as well as inflammatory skin diseases, including psoriasis, contact dermatitis and atopic dermatitis. In conclusion, luteolin might be a promising molecule for the development of topic formulations and systemic agents against inflammatory skin diseases.

78 citations


Journal ArticleDOI
TL;DR: It is suggested that luteolin intake can enhance brain insulin resistance and neuroinflammation, directly and indirectly, to protect against the development of Alzheimer's‐like disease, and the gut microbiota‐liver‐brain axis is mainly involved in the indirect pathway.
Abstract: Luteolin is a widely distributed flavone herbs and vegetables. It has anti-oxidant and anti-inflammatory activities and improves glucose metabolism by potentiating insulin sensitivity and improving β-cell function and mass. Alzheimer's disease (AD) is induced by the deposition of amyloid-beta (Aβ) in the hippocampus and the formation of neurotoxic Aβ plaques. The Aβ deposition is associated with increased formation of Aβ from amyloid precursor protein by up-regulation of β-secretase and β-site amyloid precursor protein-cleaving enzyme 1 (BACE1). Furthermore, Aβ accumulation is increased by brain insulin resistance. The impairment of insulin/IGF-1 signaling mainly in the hippocampus and brain insulin resistance is connected to signals originating in the liver and gut microbiota, known as the gut microbiota-liver-brain axis. This indicates that the changes in the production of short-chain fatty acids by the gut microbiota and pro-inflammatory cytokines can alter insulin resistance in the liver and brain. Luteolin is detected in the brain tissues after passing through the blood-brain barrier, where it can directly influence neuroinflammation and brain insulin resistance and modulate Aβ deposition. Luteolin (10-70 mg/kg bw for rodents) can modulate the systemic and brain insulin resistance, and it suppresses AD development directly, and it influences Aβ deposition by activation of the gut microbiota-liver-brain axis. In this review, we evaluate the potential of luteolin to mitigate two potential causes of AD, neuroinflammatory processes, and disruption of glucose metabolism in the brain. This review suggests that luteolin intake can enhance brain insulin resistance and neuroinflammation, directly and indirectly, to protect against the development of Alzheimer's-like disease, and the gut microbiota-liver-brain axis is mainly involved in the indirect pathway. However, most studies have been conducted in animal studies, and human clinical trials are needed.

50 citations


Journal ArticleDOI
TL;DR: A review of the anti-inflammatory, antioxidant, and immunomodulatory effects of C. longa and curcumin (CUR) can be found in this article.
Abstract: Curcuma longa (C. longa) or turmeric is a plant with a long history of use in traditional medicine, especially for treating inflammatory conditions C. longa and its main constituent, curcumin (CUR), showed various pharmacological effects such as antioxidant and anti-microbial properties. The updated knowledge of anti-inflammatory, antioxidant, and immunomodulatory effects of C. longa and CUR is provided in this review article. Pharmacological effects of C. longa, and CUR, including anti-inflammatory, antioxidant, and immunomodulatory properties, were searched using various databases and appropriate keywords until September 2020. Various studies showed anti-inflammatory effects of C. longa and CUR, including decreased white blood cell, neutrophil, and eosinophil numbers, and its protective effects on serum levels of inflammatory mediators such as phospholipase A2 and total protein in different inflammatory disorders. The antioxidant effects of C. longa and CUR were also reported in several studies. The plant extracts and CUR decreased malondialdehyde and nitric oxide levels but increased thiol, superoxide dismutase, and catalase levels in oxidative stress conditions. Treatment with C. longa and CUR also improved immunoglobulin E (Ig)E, pro-inflammatory cytokine interleukin 4 (IL)-4, transforming growth factor-beta, IL-17, interferon-gamma levels, and type 1/type 2 helper cells (Th1)/(Th2) ratio in conditions with disturbance in the immune system. Therefore C. longa and CUR showed anti-inflammatory, antioxidant, and immunomodulatory effects, indicating a potential therapeutic effect of the plant and its constituent, CUR, for treating of inflammatory, oxidative, and immune dysregulation disorders.

38 citations


Journal ArticleDOI
TL;DR: Melatonin administration has great potential to exert an anti‐inflammatory role and provide protection against obesity and ischemic stroke conditions, and the efficacy of this hormonal treatment on isChemic stroke with concomitant obesity, when more serious inflammation is generated is still lacking.
Abstract: Obesity is a predominant risk factor in ischemic stroke and is commonly comorbid with it. Pathologies following these conditions are associated with systemic and local inflammation. Moreover, there is increasing evidence that the susceptibility for ischemic brain damage increases substantially in experimental models of ischemic stroke with concomitant obesity. Herein, we explore the proinflammatory events that occur during ischemic stroke and obesity, and we discuss the influence of obesity on the inflammatory response and cerebral damage outcomes in experimental models of brain ischemia. In addition, because melatonin is a neurohormone widely reported to exhibit protective effects in various diseases, this study also demonstrates the anti-inflammatory role and possible mechanistic actions of melatonin in both epidemic diseases. A summary of research findings suggests that melatonin administration has great potential to exert an anti-inflammatory role and provide protection against obesity and ischemic stroke conditions. However, the efficacy of this hormonal treatment on ischemic stroke with concomitant obesity, when more serious inflammation is generated, is still lacking.

30 citations


Journal ArticleDOI
TL;DR: In this paper, the anti-inflammatory role of luteolin and the pathways it may act on are discussed, and the possible role of microbiota in inflammatory modulation by lutolin is discussed.
Abstract: Luteolin belongs to the family of flavonoids, which have anti-inflammatory functions, potentially useful in a clinical context, particularly for patients suffering from cancer, neuropsychiatric disorders, inflammatory bowel conditions. This peculiarity has been used for centuries in traditional Chinese medicine, for many different diseases. Its anti-inflammatory effects might be particularly relevant in cancer, with some studies reporting anti-angiogenesis, anti-metastatic, and apoptotic effects on cancer cells by luteolin and other flavonoids. In this article, we analyze the anti-inflammatory role of luteolin, discussing the pathways it may act on. We will then discuss the possible role of microbiota in inflammatory modulation by luteolin. Finally, the possible therapeutic applications of luteolin's anti-inflammatory properties will be analyzed, with a particular focus on cancer.

29 citations


Journal ArticleDOI
TL;DR: The present review suggests that phytochemicals are able to attenuate the overwhelming inflammatory responses developed during sepsis by modulating different signaling pathways, and make them potent compounds to be included as complementary therapeutic agents in the diets of patients suffering from sepsi in an effort to alleviateSepsis and its life‐threatening complications, such as multi‐organ failure.
Abstract: Sepsis and septic shock are still a leading cause of mortality and morbidity in intensive care units worldwide. Sepsis is an uncontrolled and excessive response of the innate immune system toward the invading infectious microbes, characterized by the hyper-production of pro-inflammatory mediators such as interleukin (IL)-1β, IL-6, tumor-necrosis factor (TNF)-α, and high-mobility group box 1 (HMGB1). In severe sepsis, the overwhelming production of pro-inflammatory cytokines and reactive oxygen species may compromise organ function and lead to the induction of abnormal apoptosis in different organs, resulting in multiple organ dysfunction syndrome and death. Hence, compounds that are able to attenuate inflammatory responses may have therapeutic potential for sepsis treatment. Understanding the pathophysiology and underlying molecular mechanisms of sepsis may provide useful insights in the discovery and development of new effective therapeutics. Therefore, numerous studies have invested much effort into elucidating the mechanisms involved with the onset and development of sepsis. The present review mainly focuses on the molecules and signaling pathways involved in the pathogenicity of sepsis. Additionally, several well-known natural bioactive herbal compounds and phytochemicals, which have shown protective and therapeutic effects with regard to sepsis, as well as their mechanisms of action, are presented. This review suggests that these phytochemicals are able to attenuate the overwhelming inflammatory responses developed during sepsis by modulating different signaling pathways. Moreover, the anti-inflammatory and cytoprotective activities of phytochemicals make them potent compounds to be included as complementary therapeutic agents in the diets of patients suffering from sepsis in an effort to alleviate sepsis and its life-threatening complications, such as multi-organ failure.

24 citations


Journal ArticleDOI
TL;DR: In this article, the effect of redox imbalance in non-alcoholic fatty liver disease (NAFLD) was investigated in several Phase II and Phase III trials, and the results showed that low-iron exposure induced protective effects related to nuclear factor-κB, signal transducer and activation of transcription 3, and nuclear factor erythroid-related factor 2 (Nrf2).
Abstract: Oxidative stress is an imbalance between oxidants and antioxidants in favor of the oxidants, leading to a disruption of redox signaling and control, and/or molecular damage altering cellular functions. This redox imbalance may trigger different responses depending on the antioxidant potential of a given cell, the level of reactive oxygen/nitrogen species (ROS/RNS) attained and the time of exposure, with protective effects being induced at low ROS/RNS levels in acute or short-term conditions, and harmful effects after high ROS/RNS exposure in prolonged situations. Relevant conditions underlying liver redox imbalance include iron overload associated with ROS production via Fenton chemistry and the magnitude of the iron labile pool achieved, with low iron exposure inducing protective effects related to nuclear factor-κB, signal transducer and activation of transcription 3, and nuclear factor erythroid-related factor 2 (Nrf2) activation and upregulation of ferritin, hepcidin, acute-phase response and antioxidant components, whereas high iron exposure causes drastic oxidation of biomolecules, mitochondrial dysfunction, and cell death due to necrosis, apoptosis and/or ferroptosis. Redox imbalance in nonalcoholic fatty liver disease (NAFLD) is related to polyunsaturated fatty acid depletion, lipogenic factor sterol regulatory element-binding protein-1c upregulation, fatty acid oxidation-dependent peroxisome proliferator-activated receptor-α downregulation, low antioxidant factor Nrf2 and insulin resistance, a phenomenon that is exacerbated in nonalcoholic steatohepatitis triggering an inflammatory response. Thyroid hormone (T3 ) administration determines liver preconditioning against ischemia-reperfusion injury due to the redox activation of several transcription factors, AMP-activated protein kinase, unfolded protein response and autophagy. High grade liver redox imbalance occurring in severe iron overload is adequately handled by iron chelation, however, that underlying NAFLD/NASH is currently under study in several Phase II and Phase III trials.

22 citations


Journal ArticleDOI
TL;DR: Curcumin is a plant-derived agent that has shown interesting properties for cancer therapy and has shown that not only directly inhibit the growth of cancer cells, but can also modulate the growth and activity of immunosuppressant and tumor-promoting cells as discussed by the authors.
Abstract: The tumor microenvironment (TME) is made up of several cells and molecules that affect the survival of cancer cells. Indeed, certain (immunosuppressive) cells which promote tumors can promote the growth of tumors by stimulating the proliferation of cancer cells and promoting angiogenesis. During tumor growth, antitumoral immunity includes natural killer cells and CD8+ T cells cannot overcome immunosuppressive responses and cancer cell proliferation. In order to achieve the appropriate therapeutic response, we must kill cancer cells and suppress the release of immunosuppressive molecules. The balance between anti-tumor immunity and immunosuppressive cells, such as regulatory T cells (Tregs), cancer-associated fibroblasts, tumor-associated macrophages, and myeloid-derived suppressor cells plays a key role in the suppression or promotion of cancer cells. Curcumin is a plant-derived agent that has shown interesting properties for cancer therapy. It has shown that not only directly inhibit the growth of cancer cells, but can also modulate the growth and activity of immunosuppressant and tumor-promoting cells. In this review, we explain how curcumin modulates interactions within TME in favor of tumor treatment. The potential modulating effects of curcumin on the responses of cancer cells to treatment modalities such as immunotherapy will also be discussed.

21 citations


Journal ArticleDOI
TL;DR: In this article, the authors summarized possible mechanisms by which curcumin could interfere with self-assembly processes and reduce amyloid aggregation, and discussed clinical trials in which curcurumin is used as a therapeutic agent for the treatment of diseases linking to protein aggregates.
Abstract: Amyloidosis is a concept that implicates disorders and complications that are due to abnormal protein accumulation in different cells and tissues. Protein aggregation-associated diseases are classified according to the type of aggregates and deposition sites, such as neurodegenerative disorders and type 2 diabetes mellitus. Polyphenolic phytochemicals such as curcumin and its derivatives have anti-amyloid effects both in vitro and in animal models; however, the underlying mechanisms are not understood. In this review, we summarized possible mechanisms by which curcumin could interfere with self-assembly processes and reduce amyloid aggregation in amyloidosis. Furthermore, we discuss clinical trials in which curcumin is used as a therapeutic agent for the treatment of diseases linking to protein aggregates.

Journal ArticleDOI
TL;DR: In this paper, the authors provide compelling evidence that certain flavonoids, especially luteolin formulated in olive pomace oil together with hydroxytyrosol, offers a reasonable prophylactic treatment approach due to its many beneficial actions.
Abstract: Alzheimer's disease (AD), already the world's most common form of dementia, is projected to continue increasing in prevalence over the next several decades. The current lack of understanding of the pathogenesis of AD has hampered the development of effective treatments. Historically, AD research has been predicated on the amyloid cascade hypothesis (ACH), which attributes disease progression to the build-up of amyloid protein. However, multiple clinical studies of drugs interfering with ACH have failed to show any benefit demonstrating that AD etiology is more complex than previously thought. Here we review the current literature on the emerging key role of neuroinflammation, especially activation of microglia, in AD pathogenesis. Moreover, we provide compelling evidence that certain flavonoids, especially luteolin formulated in olive pomace oil together with hydroxytyrosol, offers a reasonable prophylactic treatment approach due to its many beneficial actions.

Journal ArticleDOI
TL;DR: The role of polyphenols on the aging gut has not been fully characterized, but accumulating evidence suggests that these compounds exert selective effects on the gut microbial community as discussed by the authors, which is a promising anti-aging candidate because of their ability to modulate some of the common denominators of aging.
Abstract: Aging induces significant shifts in the composition of gut microbiota associated with decreased microbial diversity. Age-related changes in gut microbiota include a loss of commensals and an increase in disease-associated pathobionts. These alterations are accelerated by lifestyle factors, such as poor nutritional habits, physical inactivity, and medications. Given that diet is one of the main drivers shaping the gut microbiota, nutritional interventions for restoring gut homeostasis are of great importance to the overall health of older adults. Polyphenols, ubiquitously present in fruits and vegetables, have emerged as promising anti-aging candidates because of their ability to modulate some of the common denominators of aging, including gut dysbiosis. These compounds can influence the composition of the gut microbiota, and gut bacteria metabolize polyphenols into bioactive compounds that produce relevant health effects. Although the role of polyphenols on the aging gut has not been fully characterized, accumulating evidence suggests that these compounds exert selective effects on the gut microbial community. Here, we discuss the reciprocal interactions between polyphenols and gut microbiota and summarize the latest findings on the effects of polyphenols on modulating intestinal bacteria during aging.

Journal ArticleDOI
TL;DR: In this article, the authors investigated if Cadmium chloride (CdCl2 )-induced hepatic steatosis and fibrosis and the protective effect of quercetin (QUR) are mediated modulating the activity of miR-21, a known hepatic lipogenic and fibrotic miRNA.
Abstract: This study investigated if cadmium chloride (CdCl2 )-induced hepatic steatosis and fibrosis and the protective effect of quercetin (QUR) are mediated modulating the activity of miR-21, a known hepatic lipogenic and fibrotic miRNA. Male rats (n = 8/group) were divided as control, control + QUR (50 mg/kg; orally), CdCl2 (10 moml/L; drinking water), CdCl2 + miR-21 antagomir (inhibitor) (16 mg/kg/first 3 days), and CdCl2 + QUR (50 mg/kg). Treatments were conducted for 20 weeks, daily. All treatments showed no effect on fasting glucose and insulin levels. Administration of either miR-21 or QUR prevented CdCl2 -induced hepatic damage, as well as lipid droplets and collagen deposition. They also reduced serum levels of ALT and AST and decreased serum and hepatic levels of total cholesterol, triglycerides, and low-density lipoproteins in CdCl2 -treated rats. Concomitantly, they reduced hepatic levels of reactive oxygen species, malondialdehyde, interleukin-6, and tumor necrosis factor-α, suppressed the activation of NF-kb P65, and increased hepatic levels of nuclear factor erythroid 2-related factor 2 (Nrf2), glutathione (GSH), and superoxide dismutase (SOD). These effects were associated with reduced expression of SREBP1, TGF-β1, Smad3, and collagen1 A and increased expression of PPARα, CPT1, and smad7. Interestingly, QUR significantly lowered levels of miR-21 and increased the protein levels and activity of Nrf2, as well as levels of GSH and SOD in the livers of both the control and CdCl2 -treated rats. Of note, levels of Nrf2 were negatively correlated with the transcription of miR-21. In conclusion: QUR prevents CdCl2 -induced hepatic steatosis and fibrosis mainly through attenuating its ability to upregulate miR-21, at least, by upregulation of Nrf2.

Journal ArticleDOI
Yonggang Zuo1, Changping Qu1, Yanyan Tian1, Yuqing Wen1, Shuguan Xia1, Mingde Ma1 
TL;DR: In this article, a regulatory axis of HIF-1, SNHG1, miR-199a-3p, and mitochondrial transcription factor A (TFAM) involved in tumor angiogenesis and metastasis under hypoxic conditions in BC was reported.
Abstract: Activation of hypoxia-inducible factors (HIFs) as a result of intratumoral hypoxia modulates a cascade of molecular pathways thus leading to angiogenesis and metastasis in many solid tumors, including breast cancer (BC). In our paper, we report a regulatory axis of HIF-1, SNHG1, miR-199a-3p, and mitochondrial transcription factor A (TFAM) involved in tumor angiogenesis and metastasis under hypoxic conditions in BC. The expression of SNHG1 was determined in human BC cells cultured in hypoxia (1% O2 , 24 h) and normoxia (20% O2 , 24 h). Cultured MDA-MB-231 cells were assayed for the proliferation, migration, invasion, angiogenesis in vitro by using EdU staining, transwell chamber assays, Matrigel-based angiogenesis assays, tumorigenesis, and lung metastasis in vivo by using an orthotopic-transplant model of human BC. Dual-luciferase reporter assay, chromatin immunoprecipitation quantitative polymerase chain reaction assay, fluorescence in situ hybridization assay, RNA-binding protein immunoprecipitation assay, and RNA pull-down were performed to test interaction between HIF-1 and SNHG1, SNHG1 and miR-199a-3p, miR-199a-3p and TFAM. SNHG1 was increased under hypoxic conditions at a HIF-1-dependent manner. SNHG1 knockdown tempered MDA-MB-231 cell proliferation, migration, invasion, angiogenesis, in vitro, tumorigenesis, and lung metastasis in vitro. SNHG1 was co-expressed with miR-199a-3p and regulated the TFAM, a target gene of miR-199a-3p. SNHG1 increased the TFAM by binding with miR-199a-3p, thus promoting BC development and metastasis. These results support a regulatory axis consisting of HIF-1, SNHG1, miR-199a-3p, and TFAM during BC development and metastasis under hypoxic conditions, providing an opportunity to develop targeted therapeutics for BC.

Journal ArticleDOI
TL;DR: In this article, a review aims to update the current information about the modulation of the pre-established gut microbiota dysbiosis by dietary phenolic compounds in both animal studies and human trials, distinguishing the preventive or treatment approaches in animal studies.
Abstract: The human intestine contains an intricate ecological community of bacteria, referred as the gut microbiota, which plays a pivotal role in the host homeostasis. Multiple factors could interfere with this delicate balance, thus causing a disruption of the microbiota equilibrium, the so called dysbiosis. Gut microbiota dysbiosis is involved in gastrointestinal and extra-intestinal metabolic diseases, as obesity and diabetes. Polyphenols, present in a broad range of plant foods, are known to have numerous health benefits; however, their beneficial effect on pre-existing dysbiosis is less clear. Indeed, in most of the conducted animal studies the administration of polyphenols or foods rich in polyphenols occurred simultaneously with the induction of the pathology to be examined, then analyzing the preventive action of the polyphenols on the onset of dysbiosis, while very low studies analyzed the modulatory activity of polyphenols on the pre-existing dysbiosis. For this reason, the present review aims to update the current information about the modulation of the pre-established gut microbiota dysbiosis by dietary phenolic compounds in a broad range of disorders in both animal studies and human trials, distinguishing the preventive or treatment approaches in animal studies. The described studies highlight that dietary polyphenols, exerting prebiotic-like effects, can modulate the pre-existing dysbiosis stimulating the growth of beneficial bacteria and inhibiting pathogenic bacteria in both animal models and humans. Anyway, most of the conducted studies are related to obesity and metabolic syndrome, and so further studies are needed to understand this polyphenols' ability in relation to other pathologies.

Journal ArticleDOI
Jingjing Shao1, Shibo Liu1, Xiao Zheng1, Jing Chen1, Lei Li1, Zhimin Zhu1 
TL;DR: In conclusion, targeting ROS‐mediated IRS‐1 signaling, BBR acted as an efficient agent to advance osseointegration in DM, which indicated that BBR use is a good strategy for future implants restoration in diabetic patients.
Abstract: Accompanying with diabetes mellitus-induced osteoporosis (DM-OS), diabetic patients show poor peri-implant osteogenesis after implantation for dentition defect. Berberine (BBR), a candidate oral hypoglycemic agent, is a promising agent for treating DM-OS. In this study, BBR was applied on DM rats and high-glucose-cultured bone mesenchymal stem cells (BMSCs) to investigate its therapeutic mechanism on DM-OS, thus laying a theoretical basis for the future application of BBR in implant restoration. Phenotypes were assessed in the DM rats after 4 w of gavage with BBR. Furthermore, BMSCs were cultured with high glucose and BBR. Cell Counting Kit-8, 2',7'-dichlorofluorescin diacetate (H2 DCF-DA), quantitative real-time PCR (qRT-PCR), and western blot were performed to estimate the cell proliferation, oxidative stress, and osteogenic differentiation. Moreover, the DM rats treated with BBR and insulin receptor substrate-1 anti-sense oligonucleotide (IRS-1-ASO) underwent a 4-w implant-healing period and then micro computed tomography (Micro-CT) and histology were performed to verify the mechanism. Results showed that the 4-w administration of BBR markedly improved the glucose metabolism and bone metabolism in the DM rats. in vitro experiments revealed that BBR alleviated high-glucose-inhibited osteogenesis of the BMSCs by upregulating reactive oxygen species (ROS)-mediated IRS-1 signaling. Besides, injection of IRS-1-ASO abolished the BBR promotion of implant osseointegration in the DM rats. In conclusion, targeting ROS-mediated IRS-1 signaling, BBR acted as an efficient agent to advance osseointegration in DM, which indicated that BBR use is a good strategy for future implants restoration in diabetic patients.

Journal ArticleDOI
TL;DR: In this article, the authors explored the neuroprotection mediated by daidzein in hypothalamic neurons by using a membrane-based model of obesity-related neuroinflammation and found that Daidzeins protected hfHypo GnRH cells by downregulating cell death, proinflammatory processes, oxidative stress, and apoptosis.
Abstract: Phytoestrogens can control high-fat diet-induced hypothalamic inflammation that is associated with severe consequences, including obesity, type 2 diabetes, cardiovascular and neurodegenerative diseases. However, the phytoestrogen anti-neuroinflammatory action is poorly understood. In this study, we explored the neuroprotection mediated by daidzein in hypothalamic neurons by using a membrane-based model of obesity-related neuroinflammation. To test the daidzein therapeutic potential a biohybrid membrane system, consisting of hfHypo GnRH-neurons in culture on PLGA membranes, was set up. It served as reliable in vitro tool capable to recapitulate the in vivo structure and function of GnRH hypothalamic tissue. Our findings highlighted the neuroprotective role of daidzein, being able to counteract the palmitate induced neuroinflammation. Daidzein protected hfHypo GnRH cells by downregulating cell death, proinflammatory processes, oxidative stress, and apoptosis. It also restored the proper cell morphology and functionality through a mechanism which probably involves the activation of ERβ and GPR30 receptors along with the expression of GnRH peptide and KISS1R.

Journal ArticleDOI
TL;DR: In this paper, the authors reviewed the protective effects and the therapeutic role of various bioactive compounds from plants in pulmonary fibrosis management, including plant extracts and active compound that directly target the processes involved in PF, could be suitable therapeutic options with less adverse effects.
Abstract: Pulmonary fibrosis (PF) is the devastating consequence of various inflammatory diseases of the lung. PF leads to a reduction of lung function, respiratory failure, and death. Several molecular pathways are involved in PF, such as inflammatory cytokines including tumor necrosis factor α (TNFα), tumor necrosis factor β1 (TNFβ1), interleukin 6 (IL-6), and interleukin 4 (IL-4), reactive oxygen species, matrix metalloproteases, and transforming growth factor-beta (TGF-β). Targeting these processes involved in the progression of PF is essential for the treatment of this disease. Natural products, including plant extracts and active compound that directly target the processes involved in PF, could be suitable therapeutic options with less adverse effects. In the present study, we reviewed the protective effects and the therapeutic role of various bioactive compounds from plants in PF management.

Journal ArticleDOI
TL;DR: In this article, two new cytokines that suppress inflammation, IL-37 and IL-38, have been proposed, which are all anti-inflammatory molecules with different signaling pathways, and it is pertinent to recommend the combination of luteolin with these antiinflammatory cytokines in inflammation.
Abstract: Luteolin belongs to the flavone family originally present in some fruits and vegetables, including olives, which decrease intracellular levels of reactive oxygen species (ROS) following the activation of various stimuli. Luteolin inhibits inflammation, a complex process involving immune cells that accumulate at the site of infectious or non-infectious injury, with alteration of the endothelium leading to recruitment of leukocytes. Cytokines have been widely reported to act as immune system mediators, and IL-1 family members evolved to assist in host defense against infections. Interleukin (IL)-1 and Toll-like receptor (TLR) are involved in the innate immunity in almost all living organisms. After being synthesized, IL-1 induces numerous inflammatory mediators including itself, other pro-inflammatory cytokines/chemokines, and arachidonic acid products, which contribute to the pathogenesis of immune diseases. Among the 11 members of the IL-1 family, there are two new cytokines that suppress inflammation, IL-37 and IL-38. IL-38 binds IL-36 receptor (IL-1R6) and inhibits several pro-inflammatory cytokines, including IL-6, through c-Jun N-terminal kinase (JNK) induction and reducing AP1 and nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) activity, alleviating inflammatory diseases. Therefore, since luteolin, IL-37 and IL-38 are all anti-inflammatory molecules with different signaling pathways, it is pertinent to recommend the combination of luteolin with these anti-inflammatory cytokines in inflammation.

Journal ArticleDOI
TL;DR: In this paper, the effects of carvacrol and pioglitazone on the PPAR-γ receptors were investigated in rats inhaled with paraquat aerosol.
Abstract: Control rats were exposed to saline aerosol, two groups were exposed to paraquat (PQ), 27 (PQ-L) and 54 (PQ-H) mg/m3 aerosols and six groups were treated with carvacrol, 20 (C-L) and 80 (C-H) mg/kg/day, pioglitazone, 5 (Pio-L) and 10 (Pio-H) mg/kg/day, C-L+Pio-L and dexamethasone, 0.03 mg/kg/day, for 16 days after the end of exposure to PQ-H. Different variables were measured after the end of treatment period. Total and differential white blood cells counts, nitrite, malondialdehyde, interleukin (IL)-10, and interferon-gamma levels were significant increased, but thiol, superoxide dismutase, catalase, IL-17, and tumor necrosis factor alpha were decreased in the blood due to both doses of PQ (p < 0.05-p < 0.001). Most measured parameters were significantly improved in treated groups with both doses of carvacrol, pioglitazone, the combination of C-L+Pio-L and dexamethasone compared to PQ-H group (p < 0.05-p < 0.001). Treatment with C-L+Pio-L showed significantly higher effects compared to each one alone (p < 0.05-p < 0.001). Systemic oxidative stress and inflammation due to inhaled PQ were improved by carvacrol and pioglitazone. Higher effects of C-L+Pio-L than each one alone suggests carvacrol modulating PPAR-γ receptors.

Journal ArticleDOI
TL;DR: In this article, the authors focused on targeting the lactate metabolism/transporters, and lactate-associated mediators, including glycolytic pathways, for preventing and managing tumorigenesis.
Abstract: Anticancer drugs are not purely effective because of their toxicity, side effects, high cost, inaccessibility, and associated resistance. On the other hand, cancer is a complex public health problem that could intelligently adopt different signaling pathways and alter the body's metabolism to escape from the immune system. One of the cancer strategies to metastasize is modifying pH in the tumor microenvironment, ranging between 6.5 and 6.9. As a powerful determiner, lactate is responsible for this acidosis. It is involved in immune stimulation, including innate and adaptive immunity, apoptotic-related factors (Bax/Bcl-2, caspase), and glycolysis pathways (e.g., GLUT-1, PKM2, PFK, HK2, MCT-1, and LDH). Lactate metabolism, in turn, is interconnected with several dysregulated signaling mediators, including PI3K/Akt/mTOR, AMPK, NF-κB, Nrf2, JAK/STAT, and HIF-1α. Because of lactate's emerging and critical role, targeting lactate production and its transporters is important for preventing and managing tumorigenesis. Hence, exploring and developing novel promising anticancer agents to minimize human cancers is urgent. Based on numerous studies, natural secondary metabolites as multi-target alternative compounds with health-promoting properties possess more high effectiveness and low side effects than conventional agents. Besides, the mechanism of multi-targeted natural sources is related to lactate production and cancer-associated cross-talked factors. This review focuses on targeting the lactate metabolism/transporters, and lactate-associated mediators, including glycolytic pathways. Besides, interconnected mediators to lactate metabolism are also targeted by natural products. Accordingly, plant-derived secondary metabolites are introduced as alternative therapies in combating cancer through modulating lactate metabolism and glycolytic pathways.

Journal ArticleDOI
TL;DR: In this paper, a more in-depth analysis of CoQ10 secondary deficiency was undertaken to consider its origins, introduce a new way of classification, and include aging as a form of secondary deficiency.
Abstract: Coenzyme Q10 (CoQ10 ) deficiency is a rare disease characterized by a decreased accumulation of CoQ10 in cell membranes. Considering that CoQ10 synthesis and most of its functions are carried out in mitochondria, CoQ10 deficiency cases are usually considered a mitochondrial disease. A relevant feature of CoQ10 deficiency is that it is the only mitochondrial disease with a successful therapy available, the CoQ10 supplementation. Defects in components of the synthesis machinery caused by mutations in COQ genes generate the primary deficiency of CoQ10 . Mutations in genes that are not directly related to the synthesis machinery cause secondary deficiency. Cases of CoQ10 deficiency without genetic origin are also considered a secondary deficiency. Both types of deficiency can lead to similar clinical manifestations, but the knowledge about primary deficiency is deeper than secondary. However, secondary deficiency cases may be underestimated since many of their clinical manifestations are shared with other pathologies. This review shows the current state of secondary CoQ10 deficiency, which could be even more relevant than primary deficiency for clinical activity. The analysis covers the fundamental features of CoQ10 deficiency, which are necessary to understand the biological and clinical differences between primary and secondary CoQ10 deficiencies. Further, a more in-depth analysis of CoQ10 secondary deficiency was undertaken to consider its origins, introduce a new way of classification, and include aging as a form of secondary deficiency.

Journal ArticleDOI
TL;DR: The emerging evidence has shown that mangiferin can improve renal interstitial fibrosis, pulmonary Fibrosis, myocardial fibrosis and hepatic fibrosis through the inhibition of inflammation, oxidative stress and fibrogenesis effects, indicating that Mangiferin is promising therapeutic choice for organ fibrosis.
Abstract: Fibrosis is the end stage of many chronic diseases, which results in organ function failure and high mortality. Mangiferin is a major constituent in mango and other 16 plants, and has been shown a variety of pharmacological effects, such as antioxidant, antibacterial, anti-tumor, anti-inflammation. The emerging evidence has shown that mangiferin can improve renal interstitial fibrosis, pulmonary fibrosis, myocardial fibrosis and hepatic fibrosis through the inhibition of inflammation, oxidative stress and fibrogenesis effects, indicating that mangiferin is promising therapeutic choice for organ fibrosis. The aim of this review is to summarize the therapeutic effects of mangiferin on fibrosis of various organs and the underlying mechanisms.

Journal ArticleDOI
TL;DR: In this paper, the authors reported that the induction of ferroptosis in activated stellate cells was required for dihydroartemisinin (DHA) to alleviate hepatic fibrosis.
Abstract: Targeting the elimination of activated hepatic stellate cells (HSCs) and blocking excessive deposition of extracellular matrix are recognized as an effective strategy for the treatment of hepatic fibrosis. As a newly discovered programmed cell death mode, the regulatory mechanism of ferroptosis in the clearance of activated HSCs has not been fully elucidated. In the present study, we reported that the induction of ferroptosis in activated HSCs was required for dihydroartemisinin (DHA) to alleviate hepatic fibrosis. Treatment with DHA could improve the damage of hepatic fibrosis in vivo and inhibit the activation of HSCs in vitro. Interestingly, DHA treatment could trigger ferroptosis to eliminate activated HSCs characterized by iron overload, lipid ROS accumulation, glutathione depletion, and lipid peroxidation. Specific ferroptosis inhibitors ferrostatin-1 and liproxstatin-1 could impair DHA-induced ferroptosis and also damage DHA-mediated the inhibition of activated HSCs. Importantly, autophagy activation may be closely related to DHA-induced ferroptosis. ATG5 siRNA could prevent DHA-mediated autophagy activation and ferroptosis induction, whereas ATG5 plasmid could promote the effect of DHA on autophagy and ferroptosis. Of note, the upregulation of nuclear receptor coactivator 4 (NCOA4) may play a critical role in the molecular mechanism. NCOA4 siRNA could impair DHA-induced ferroptosis, whereas NCOA4 plasmid could enhance the promoting effect of DHA on ferroptosis. Overall, our study revealed the potential mechanism of DHA against hepatic fibrosis and showed that ferroptosis could be a new way to eliminate activated HSCs.

Journal ArticleDOI
TL;DR: Stem cells' pivotal roles in promoting angiogenesis and consequent improved cardiac healing and remodeling processes should not be ignored, especially in the case of stem cell‐derived extracellular vesicles.
Abstract: Finding effective treatments for cardiac diseases is among the hottest subjects in medicine; cell-based therapies have brought great promises for managing a broad range of life-threatening heart complications such as myocardial infarction. After clarifying the critical role of angiogenesis in tissue repair and regeneration, various stem/progenitor cell were utilized to accelerate the healing of injured cardiac tissue. Embryonic, fetal, adult, and induced pluripotent stem cells have shown the appropriate proangiogenic potential for tissue repair strategies. The capability of stem cells for differentiating into endothelial lineages was initially introduced as the primary mechanism involved in improving angiogenesis and accelerated heart tissue repair. However, recent studies have demonstrated the leading role of paracrine factors secreted by stem cells in advancing neo-vessel formation. Genetically modified stem cells are also being applied for promoting angiogenesis regarding their ability to considerably overexpress and secrete angiogenic bioactive molecules. Yet, conducting further research seems necessary to precisely identify molecular mechanisms behind the proangiogenic potential of stem cells, including the signaling pathways and regulatory molecules such as microRNAs. In conclusion, stem cells' pivotal roles in promoting angiogenesis and consequent improved cardiac healing and remodeling processes should not be ignored, especially in the case of stem cell-derived extracellular vesicles.

Journal ArticleDOI
TL;DR: Overall, while the research on SPMs is still evolving, they may offer a novel therapeutic option for inflammatory conditions and suggest they may be a valuable resource in the fight against Coronavirus disease‐19 (COVID‐19).
Abstract: Specialized proresolving mediators (SPMs) are endogenous lipid metabolites of long-chain polyunsaturated fatty acids that are involved in promoting the resolution of inflammation. Many disease conditions characterized by excessive inflammation have impaired or altered SPM biosynthesis, which may lead to chronic, unresolved inflammation. Exogenous administration of SPMs in infectious conditions has been shown to be effective at improving infection clearance and survival in preclinical models. SPMs have also shown tremendous promise in the context of inflammatory lung conditions, such as acute respiratory distress syndrome and chronic obstructive pulmonary disease, mostly in preclinical settings. To date, SPMs have not been studied in the context of the novel Coronavirus, severe acute respiratory syndrome Coronavirus-2 (SARS-CoV-2), however their preclinical efficacy in combatting infections and improving acute respiratory distress suggest they may be a valuable resource in the fight against Coronavirus disease-19 (COVID-19). Overall, while the research on SPMs is still evolving, they may offer a novel therapeutic option for inflammatory conditions.

Journal ArticleDOI
TL;DR: This article showed that prolonged fasting in catalase-knockout (KO) mice drastically increased ROS production, which induced liver-specific pexophagy, an autophagic degradation of peroxisomes.
Abstract: Peroxisomes are dynamic organelles that participate in a diverse array of cellular processes, including β-oxidation, which produces a considerable amount of reactive oxygen species (ROS). Although we showed that catalase depletion induces ROS-mediated pexophagy in cells, the effect of catalase deficiency during conditions that favor ROS generation remains elusive in mice. In this study, we reported that prolonged fasting in catalase-knockout (KO) mice drastically increased ROS production, which induced liver-specific pexophagy, an autophagic degradation of peroxisomes. In addition, increased ROS generation induced the production of pro-inflammatory cytokines in the liver tissues of catalase-KO mice. Furthermore, there was a significant increase in the levels of aspartate transaminase and alanine transaminase as well as apparent cell death in the liver of catalase-KO mice during prolonged fasting. However, an intra-peritoneal injection of the antioxidant N-acetyl-l-cysteine (NAC) and autophagy inhibitor chloroquine inhibited the inflammatory response, liver damage, and pexophagy in the liver of catalase-KO mice during prolonged fasting. Consistently, genetic ablation of autophagy, Atg5 led to suppression of pexophagy during catalase inhibition by 3-aminotriazole (3AT). Moreover, treatment with chloroquine also ameliorated the inflammatory response and cell death in embryonic fibroblast cells from catalase-KO mice. Taken together, our data suggest that ROS-mediated liver-specific pexophagy observed during prolonged fasting in catalase-KO mice may be responsible for the process associated with hepatic cell death.

Journal ArticleDOI
TL;DR: In this paper, an LncRNA associated with FAS translational regulation (lnc-AFTR) through exosomal RNA sequencing was shown to have anti-inflammatory and anti-apoptotic effects by inhibiting the activation of TNF signaling pathway and mitogen-activated protein kinases (MAPK) pathway.
Abstract: Although the specific expression of long noncoding RNA (lncRNA) in mastitis tissue has been reported, few studies have involved the differential expression of lncRNA in mastitis exosomes (Exo) and its mechanism and function. We screened an lncRNA associated with FAS translational regulation (lnc-AFTR) through exosomal RNA sequencing, and clarified its function and molecular mechanism. Lnc-AFTR is markedly downregulated in Staphylococcus aureus-Exo and S. aureus-induced MAC-T cell as well as mastitis tissue. Overexpression of lnc-AFTR exosomes (oe-AFTR-Exo) significantly improves cell damage induced by S. aureus, including inhibiting apoptosis, promoting proliferation, and increasing the production of pro-inflammatory cytokines (tumor necrosis factor-α [TNF-α] and interleukin-1β [IL-1β]). Oe-AFTR-Exo also suppressed the activation of Caspase-8, Caspase-3, and JNK. Dual-luciferase report analysis confirmed that lnc-AFTR interacts with FAS mRNA directly to hinder translation process, but does not degrade FAS mRNA. Overexpression of lnc-AFTR in MAC-T cells obviously reduced S. aureus-induced apoptosis and inflammation. Knockdown of lnc-AFTR significantly increased FAS and promoted the activation of Caspase-8, Caspase-3, and JNK caused by S. aureus. In summary, these results revealed the mechanism by which lnc-AFTR directly bound FAS mRNA to prevent translation, and confirmed that the exosomal lnc-AFTR exerted anti-inflammatory and anti-apoptotic effects by inhibiting the activation of TNF signaling pathway and mitogen-activated protein kinases (MAPK) signaling pathway.

Journal ArticleDOI
TL;DR: In this article, the authors explored whether Se deficiency can cause ER stress and induce apoptosis in swine small intestine and established the Se deficiency swine model in vivo and the intestinal epithelial (IPEC-J2) cell Se deficiency model in vitro.
Abstract: Selenium (Se) plays a crucial role in intestinal health. However, the specific mechanism by which deficiency of Se causes intestinal damage remains unclear. This study was to explore whether Se deficiency can cause ER stress and induce apoptosis in swine small intestine. We established the Se deficiency swine model in vivo and the intestinal epithelial (IPEC-J2) cell Se deficiency model in vitro. The results of morphological observation showed that Se deficiency caused structural damage in intestinal villi and the decrease of goblet cell structure. The apoptotic characteristics such as nucleolar condensation, mitochondrial swelling, and apoptotic bodies were observed in the IPEC-J2 cells. The results of acridine orange/ethidium bromide and mitochondrial membrane potential fluorescence staining in vitro showed that there were more apoptotic cells in the Se-deficiency group than that in the control group. The protein and/or mRNA expression levels of Bax, Bcl-2, caspase 3, caspase 8, caspase 9, cytc, PERK, ATF6, IRE, XBP1, CHOP, GRP78, which are related to ER stress-apoptosis pathway, were significantly increased in the Se-deficient group which compared with the control group in vivo and in vitro were consistent. These results indicated that Se deficiency induced ER stress and increased the apoptosis in swine small intestine and IPEC-J2 cells and then caused the damage in swine small intestinal tissue. Besides, the results of gene expressions in our experiment proved that ER stress induced by Se deficiency promoted apoptosis. These results filled the blank in the mechanism of Se deficiency-induced intestinal injury in swine.