Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Biomed Pharmacother ; 172: 116202, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38330707

ABSTRACT

Hypoxia is a common feature of diabetic tissues, which highly correlates to the progression of diabetes. The formation of hypoxic context is induced by disrupted oxygen homeostasis that is predominantly driven by vascular remodeling in diabetes. While different types of vascular impairments have been reported, the specific features and underlying mechanisms are yet to be fully understood. Under hypoxic condition, cells upregulate hypoxia-inducible factor-1α (HIF-1α), an oxygen sensor that coordinates oxygen concentration and cell metabolism under hypoxic conditions. However, diabetic context exploits this machinery for pathogenic functions. Although HIF-1α protects cells from diabetic insult in multiple tissues, it also jeopardizes cell function in the retina. To gain a deeper understanding of hypoxia in diabetic complications, we focus on the formation of tissue hypoxia and the outcomes of HIF-1α dysregulation under diabetic context. Hopefully, this review can provide a better understanding on hypoxia biology in diabetes.


Subject(s)
Diabetes Mellitus , Humans , Hypoxia/complications , Retina , Vascular Remodeling , Oxygen
2.
Oxid Med Cell Longev ; 2023: 4365895, 2023.
Article in English | MEDLINE | ID: mdl-36778206

ABSTRACT

Testes produce sperms, and gamete generation relies on a proper niche environment. The disruption of hierarchical regulatory homeostasis in Leydig or Sertoli cells may evoke a sterile phenotype in humans. In this study, we recapitulated type 2 diabetes mellitus by using a high-fat diet- (HFD-) fed mouse model to identify the phenotype and potential mechanism of diabetes-induced testicular impairment. At the end of the study, blood glucose levels, testosterone structure, testicular antioxidant capacity, and testosterone level and the expression of hypoxia-inducible factor- (HIF-) 1α, apoptosis-related protein cleaved-caspase3, and autophagy-related proteins such as LC3I/II, p62, and Beclin1 were evaluated. We found that long-term HFD treatment causes the development of diabetes mellitus, implicating increased serum glucose level, cell apoptosis, and testicular atrophy (P < 0.05 vs. Ctrl). Mechanistically, the results showed enhanced expression of HIF-1α in both Sertoli and Leydig cells (P < 0.05 vs. Ctrl). Advanced glycation end products (AGEs) were demonstrated to be a potential factor leading to HIF-1α upregulation in both cell types. In Sertoli cells, high glucose treatment had minor effects on Sertoli cell autophagy. However, AGE treatment stagnated the autophagy flux and escalated cell apoptosis (P < 0.05 vs. Ctrl+Ctrl). In Leydig cells, high glucose treatment was adequate to encumber autophagy induction and enhance oxidative stress. Similarly, AGE treatment facilitated HIF-1α expression and hampered testosterone production (P < 0.05 vs. Ctrl+Ctrl). Overall, these findings highlight the dual effects of diabetes on autophagy regulation in Sertoli and Leydig cells while imposing oxidative stress in both cell types. Furthermore, the upregulation of HIF-1α, which could be triggered by AGE treatment, may negatively affect both cell types. Together, these findings will help us further understand the molecular mechanism of diabetes-induced autophagy dysregulation and testicular impairment, enriching the content of male reproductive biology in diabetic patients.


Subject(s)
Diabetes Mellitus, Type 2 , Testis , Mice , Animals , Humans , Male , Oxidative Stress , Autophagy , Testosterone , Glucose/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/pharmacology
3.
Biomed Pharmacother ; 157: 114004, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36375308

ABSTRACT

When the production of reactive oxygen species (ROS) is overloaded surpassing the capacity of the reductive rheostat, mammalian cells undergo a series of oxidative damage termed oxidative stress (OS). This phenomenon is ubiquitously detected in many human pathological conditions. Wound healing program implicates continuous neovascularization, cell proliferation, and wound remodeling. Increasing evidence indicates that reactive oxygen species (ROS) have profound impacts on the wound healing process through regulating a series of the physiological and pathological program including inflammatory response, cell proliferation, angiogenesis, granulation as well as extracellular matrix formation. In most pathological wound healing processes, excessive ROS exerts a negative role on the wound healing process. Interestingly, the moderate increase of ROS levels is beneficial in killing bacteria at the wound site, which creates a sterile niche for revascularization. In this review, we discussed the physiological rhythms of wound healing and the role of ROS in this progress, aim to explore the potential manipulation of OS as a promising therapeutic avenue.


Subject(s)
Oxidative Stress , Wound Healing , Animals , Humans , Reactive Oxygen Species , Wound Healing/physiology , Cell Proliferation , Neovascularization, Pathologic , Mammals
4.
ACS Appl Mater Interfaces ; 14(37): 42541-42557, 2022 Sep 21.
Article in English | MEDLINE | ID: mdl-36094305

ABSTRACT

Nanotechnology has shown a revolution in cancer treatments, including breast cancers. However, there remain some challenges and translational hurdles. Surgery, radiotherapy, and chemotherapy are the primary treatment methods for breast cancer, although drug combinations showed promising results in preclinical studies. Herein we report the development of a smart drug delivery system (DDS) to efficiently treat breast cancer by pyroptosis-starvation-chemotherapeutic combination. Cancer-starvation agent glucose oxidase was chemically attached to synthesized iron oxide nanoparticles which were entrapped inside poly(lactic-co-glycolic acid) along with apoptosis-associated speck-like protein containing a caspase recruitment domain plasmid and paclitaxel (PTX). An emulsion solvent evaporation method was used to prepare the DDS. The surface of the DDS was modified with chitosan to which aptamer was attached to achieve site-specific targeting. Hence, the prepared DDS could be targeted to a tumor site by both external magnet and aptamer to obtain an enhanced accumulation of drugs at the tumor site. The final size of the aptamer-decorated DDS was less than 200 nm, and the encapsulation efficiency of PTX was 76.5 ± 2.5%. Drug release from the developed DDS was much higher at pH 5.5 than at pH 7.4, ensuring the pH sensitivity of the DDS. Due to efficient dual targeting of the DDS, in vitro viability of 4T1 cells was reduced to 12.1 ± 1.6%, whereas the nontargeted group and free PTX group could reduce the viability of cells to 29.2 ± 2.4 and 46.2 ± 1.6%, respectively. Our DDS showed a synergistic effect in vitro and no severe side effects in vivo. This DDS has strong potential to treat various cancers.


Subject(s)
Breast Neoplasms , Chitosan , Nanoparticles , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Chitosan/therapeutic use , Drug Delivery Systems/methods , Emulsions , Female , Glucose Oxidase/therapeutic use , Humans , Magnetic Phenomena , Nanoparticles/chemistry , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Polylactic Acid-Polyglycolic Acid Copolymer/therapeutic use , Polymers/chemistry , Pyroptosis , Solvents
5.
Dev Biol ; 485: 61-69, 2022 05.
Article in English | MEDLINE | ID: mdl-35283102

ABSTRACT

Epigenetic regulation of gene expression plays a central role in bladder urothelium development and maintenance. ATPase-dependent chromatin remodeling is a major epigenetic regulatory mechanism, but its role in the bladder has not been explored. Here, we show the functions of Arid1a, the largest subunit of the SWI/SNF or BAF chromatin remodeling ATPase complex, in embryonic and adult bladder urothelium. Knockout of Arid1a in urothelial progenitor cells significantly increases cell proliferation during bladder development. Deletion of Arid1a causes ectopic cell proliferation in the terminally differentiated superficial cells in adult mice. Consistently, gene-set enrichment analysis of differentially expressed genes demonstrates that the cell cycle-related pathways are significantly enriched in Arid1a knockouts. Gene-set of the polycomb repression complex 2 (PRC2) pathway is also enriched, suggesting that Arid1a antagonizes the PRC2-dependent epigenetic gene silencing program in the bladder. During acute cyclophosphamide-induced bladder injury, Arid1a knockouts develop hyperproliferative and hyperinflammatory phenotypes and exhibit a severe loss of urothelial cells. A Hallmark gene-set of the oxidative phosphorylation pathway is significantly reduced in Aria1a mutants before injury and is unexpectedly enriched during injury response. Together, this study uncovers functions of Arid1a in both bladder progenitor cells and the mature urothelium, suggesting its critical roles in urothelial development and regeneration.


Subject(s)
Urinary Bladder , Urothelium , Adenosine Triphosphatases/genetics , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Mice , Mice, Knockout , Polycomb Repressive Complex 2/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Urinary Bladder/metabolism , Urothelium/metabolism
6.
Front Pharmacol ; 12: 628950, 2021.
Article in English | MEDLINE | ID: mdl-34497508

ABSTRACT

Diabetic nephropathy (DN) is a common diabetes associated complication. Thus, it is important to understand the pathological mechanism of DN and find the appropriate therapeutic strategy for it. Dl-3-n-Butylphthalide (DL-NBP) has anti-inflammatory and antioxidant effects, and been widely used for the treatment of stroke and cardiovascular diseases. In this study, we selected three different doses (20, 60, and 120 mg⋅kg-1 d-1) of DL-NBP and attempted to elucidate its role and molecular mechanism underlying DN. We found that DL-NBP, especially at the dose of 60 or 120 mg⋅kg-1 d-1, could significantly ameliorate diabetes-induced elevated blood urea nitrogen (BUN) and creatinine level, and alleviate renal fibrosis. Additionally, the elevated expressions of collagen and α-smooth muscle actin (α-SMA) in the kidney from db/db mice were found to be significantly suppressed after DL-NBP treatment. Furthermore, mechanistic studies revealed that DL-NBP inhibits pro-inflammatory cytokine levels, thereby ameliorating the development of renal fibrosis. Moreover, we found that DL-NBP could not only reduce the endoplasmic reticulum stress (ERS), but also suppress activation of the renin-angiotensin system to inhibit vascular endothelial growth factor (VEGF) level, which subsequently reduces the podocyte apoptosis in kidney of db/db mice. In a word, our findings suggest that DL-NBP may be a potential therapeutic drug in the treatment of DN.

7.
Oxid Med Cell Longev ; 2021: 1764929, 2021.
Article in English | MEDLINE | ID: mdl-34512862

ABSTRACT

The increase of oxidative stress is one of the important characteristics of mammalian luteal regression. Previous investigations have revealed the essential role of reactive oxygen species (ROS) in luteal cell death during luteolysis, while it is unknown how ROS is regulated in this process. Considering the decrease of blood flow and increase of PGF2α during luteolysis, we hypothesized that the HIF-1α pathway may be involved in the regulation of ROS in the luteal cell of the late corpus luteum (CL). Here, by using a pseudopregnant rat model, we showed that the level of both HIF-1α and its downstream BNIP3 was increased during luteal regression. Consistently, we observed the increase of autophagy level during luteolysis, which is regulated in a Beclin1-independent manner. Comparing with early (Day 7 of pseudopregnancy) and middle CL (Day 14), the level of ROS was significantly increased in late CL, indicating the contribution of oxidative stress in luteolysis. Inhibition of HIF-1α by echinomycin (Ech), a potent HIF-1α inhibitor, ameliorated the upregulation of BNIP3 and NIX, as well as the induction of autophagy and the accumulation of ROS in luteal cells on Day 21 of pseudopregnancy. Morphologically, Ech treatment delayed the atrophy of the luteal structure at the late-luteal stage. An in vitro study indicated that inhibition of HIF-1α can also attenuate PGF2α -induced ROS and luteal cell apoptosis. Furthermore, the decrease of cell apoptosis can also be observed by ROS inhibition under PGF2α treatment. Taken together, our results indicated that HIF-1α signaling is involved in the regression of CL by modulating ROS production via orchestrating autophagy. Inhibition of HIF-1α could obviously hamper the apoptosis of luteal cells and the process of luteal regression.


Subject(s)
Corpus Luteum/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Reactive Oxygen Species/metabolism , Animals , Female , Luteolysis/metabolism , Pregnancy , Pseudopregnancy , Rats , Rats, Sprague-Dawley
8.
Biomed Pharmacother ; 139: 111667, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34243608

ABSTRACT

Interactions between vascular smooth muscle cells (VSMCs), endothelial cells (ECs), pericytes (PCs) and macrophages (MФ), the major components of blood vessels, play a crucial role in maintaining vascular structural and functional homeostasis. Low-density lipoprotein (LDL) receptor-related protein-1 (LRP1), a transmembrane receptor protein belonging to the LDL receptor family, plays multifunctional roles in maintaining endocytosis, homeostasis, and signal transduction. Accumulating evidence suggests that LRP1 modulates vascular homeostasis mainly by regulating vasoactive substances and specific intracellular signaling pathways, including the plasminogen activator inhibitor 1 (PAI-1) signaling pathway, platelet-derived growth factor (PDGF) signaling pathway, transforming growth factor-ß (TGF-ß) signaling pathway and vascular endothelial growth factor (VEGF) signaling pathway. The aim of the present review is to focus on recent advances in the discovery and mechanism of vascular homeostasis regulated by LRP1-dependent signaling pathways. These recent discoveries expand our understanding of the mechanisms controlling LRP1 as a target for studies on vascular complications.


Subject(s)
Homeostasis/physiology , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Muscle, Smooth, Vascular/metabolism , Signal Transduction/physiology , Animals , Endocytosis/physiology , Endothelial Cells/metabolism , Humans , Lipoproteins, LDL/metabolism , Myocytes, Smooth Muscle/metabolism
9.
Colloids Surf B Biointerfaces ; 205: 111899, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34098363

ABSTRACT

Intracellular bacterial infection is underlying many serious human disorders, leading to high morbidity and mortality. The development of safe and efficient therapeutic agents is the most effective solutions to combat intracellular bacterial infections. Recently, ultrasmall gold nanoclusters (AuNCs) have emerged as an innovative nanoantibiotics against multidrug-resistant bacterial infections due to their inherent antibacterial activity. However, the therapeutic effects of AuNCs on intracellular bacterial infections and their effects on host cells still remain unvisited. Here, we demonstrate the therapeutic potential of 4,6-diamino-2-mercaptopyrimidine-functionalized AuNCs (AuDAMP) for intracellular multidrug-resistant infections in a co-culture model of macrophages and methicillin-resistant Staphylococcus aureus (MRSA). The AuNCs were found to show a superior intracellular antibacterial capability, which can eliminate most of the MRSA phagocytosed by macrophages, and without exhibiting obvious cytotoxicity on host RAW 264.7 macrophages at tested concentrations. More importantly, treatment of AuDAMP exerts critical roles on enhancing the innate immune response to defend against pathogens invading inside the host cells and alleviating the bacterial infection-induced inflammatory response to avoid pyroptosis by up-regulating significantly xenophagy level in macrophages. Taken together, our results suggest that AuNCs hold great potential for the treatment of intracellular bacterial infections.


Subject(s)
Bacterial Infections , Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bacterial Infections/drug therapy , Drug Resistance, Multiple, Bacterial , Gold/pharmacology , Humans , Immunity, Cellular , Microbial Sensitivity Tests , Staphylococcal Infections/drug therapy
10.
J Cell Mol Med ; 25(10): 4776-4785, 2021 05.
Article in English | MEDLINE | ID: mdl-33788387

ABSTRACT

As a serious metabolic disease, diabetes causes series of complications that seriously endanger human health. The liver is a key organ for metabolizing glucose and lipids, which substantially contributes to the development of insulin resistance and type 2 diabetes mellitus (T2DM). Exogenous fibroblast growth factor 1 (FGF1) has a great potential for the treatment of diabetes. Receptor of advanced glycation end products (RAGE) is a receptor for advanced glycation end products that involved in the development of diabetes-triggered complications. Previous study has demonstrated that FGF1 significantly ameliorates diabetes-mediated liver damage (DMLD). However, whether RAGE is involved in this process is still unknown. In this study, we intraperitoneally injected db/db mice with 0.5 mg/kg FGF1. We confirmed that FGF1 treatment not only significantly ameliorates diabetes-induced elevated apoptosis in the liver, but also attenuates diabetes-induced inflammation, then contributes to ameliorate liver dysfunction. Moreover, we found that diabetes triggers the elevated RAGE in hepatocytes, and FGF1 treatment blocks it, suggesting that RAGE may be a key target during FGF1 treatment of diabetes-induced liver injury. Thus, we further confirmed the role of RAGE in FGF1 treatment of AML12 cells under high glucose condition. We found that D-ribose, a RAGE agonist, reverses the protective role of FGF1 in AML12 cells. These findings suggest that FGF1 ameliorates diabetes-induced hepatocyte apoptosis and elevated inflammation via suppressing RAGE pathway. These results suggest that RAGE may be a potential therapeutic target for the treatment of DMLD.


Subject(s)
Acute Lung Injury/drug therapy , Diabetes Complications/drug therapy , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 2/complications , Fibroblast Growth Factor 1/pharmacology , Inflammation/drug therapy , Receptor for Advanced Glycation End Products/antagonists & inhibitors , Acute Lung Injury/etiology , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Animals , Apoptosis , Diabetes Complications/etiology , Diabetes Complications/metabolism , Diabetes Complications/pathology , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL
11.
Front Cell Dev Biol ; 9: 631016, 2021.
Article in English | MEDLINE | ID: mdl-33553188

ABSTRACT

Owing to the avascular structure of the ovarian follicle, proliferation of granulosa cells (GCs) and development of follicles occur under hypoxia, which is obviously different from the cell survival requirements of most mammalian cells. We hypothesized that autophagy may exert an inhibitory effect on GC apoptosis. To decipher the underlying mechanism, we constructed a rat follicular development model using pregnant mare serum gonadotropin and a cell culture experiment in hypoxic conditions (3% O2). The present results showed that the autophagy level was obviously increased and was accompanied by the concomitant elevation of hypoxia inducible factor (HIF)-1α and BNIP3 (Bcl-2/adenovirus E1B 19kDa-interacting protein 3) in GCs during follicular development. The levels of Bax (Bcl2-associated X) and Bcl-2 (B-cell lymphoma-2) were increased, while the activation of caspase-3 exhibited no obvious changes during follicular development. However, inhibition of HIF-1α attenuated the increase in Bcl-2 and promoted the increase in Bax and cleaved caspase-3. Furthermore, we observed the downregulation of BNIP3 and the decrease in autophagy after treatment with a specific HIF-1α activity inhibitor (echinomycin), indicating that HIF-1α/BNIP3 was involved in autophagy regulation in GCs in vivo. In an in vitro study, we also found that hypoxia did not obviously promote GC apoptosis, while it significantly enhanced the activation of HIF-1α/BNIP3 and the induction of autophagy. Expectedly, this effect could be reversed by 3-methyladenine (3-MA) treatment. Taken together, these findings demonstrated that hypoxia drives the activation of HIF-1α/BNIP3 signaling, which induces an increase in autophagy, protecting GC from apoptosis during follicular development.

12.
Carbohydr Polym ; 247: 116669, 2020 Nov 01.
Article in English | MEDLINE | ID: mdl-32829797

ABSTRACT

A quick control of heavy hemorrhaging is critical to save the life of injured person. Herein we developed a hemostat of chitosan@calcium alginate microspheres through combination of microemulsion, polyelectrolyte complexation coating, and thermally induced phase separation. The alginate coated microspheres featured a structure of a porous chitosan core and a compact calcium alginate shell layer. Their in vitro and in vivo hemostatic properties were evaluated by the whole blood clotting kinetics, and rat tail amputation and liver laceration models. Compared to the porous chitosan microspheres, the alginate coated microspheres showed much enhanced hemostatic efficiency. The latter formed bigger blood clots; its hemostatic time on the liver laceration was substantially shortened to 53 ± 10 s from 107 ± 9 s of the former. It was shown that calcium ions released from the calcium alginate layer may accelerate the formation of blood clots. Such a biocompatible microsphere is a promising quick hemostat for controlling traumatic bleeding.


Subject(s)
Alginates/chemistry , Blood Coagulation/drug effects , Chitosan/chemistry , Hemorrhage/drug therapy , Hemostasis/drug effects , Hemostatics/pharmacology , Microspheres , Animals , Hemorrhage/blood , Hemorrhage/diagnosis , Porosity , Rats , Rats, Sprague-Dawley
13.
Front Cell Dev Biol ; 8: 619924, 2020.
Article in English | MEDLINE | ID: mdl-33537309

ABSTRACT

During the luteinization after ovulation in mammalian ovary, the containing cells undergo an energy consuming function re-determination process to differentiate into luteal cells under avascular environment. Previous evidences have delineated the contribution of autophagy to the cell differentiation and the catabolic homeostasis in various types of mammalian cells, whereas few interest had been focused on the involvement of autophagy in the luteinization of granulosa cells during the formation of early corpus luteum. Herein, the present study investigated that expression and contribution of autophagy during granulosa cell luteinization and early luteal development through in vivo and in vitro experiments. The results clearly demonstrated that HIF-1α/BNIP3-mediated autophagy plays a vital role in the luteinization of granulosa cells during the early luteal formation in vivo and in vitro. In the neonatal corpus luteum, HIF-1α up-regulated BNIP3 expressions, which contributed to the autophagic initiation by disrupting beclin1 from Bcl-2/beclin1 complex and protected cells from apoptosis by curbing the skew of mitochondria balance under avascular niche. Notably, Inhibition of HIF-1α activity by echinomycin enhanced the levels of cytoplasmic cytochrome c and cell apoptosis in the nascent corpus luteum. These findings revealed that HIF-1α/BNIP3-mediated autophagy enabled the process of granulosa cell luteinization and protected the granulosa-lutein cells from further apoptosis under hypoxia niche. To our knowledge, the present study firstly clarified that HIF-1α/BNIP3-mediated autophagy contributes to the luteinization of granulosa cells during the formation of pregnant corpus luteum, which will help us further understanding the luteal biology and provide us new clues for the treatment of luteal insufficiency.

14.
Exp Ther Med ; 17(5): 4205-4212, 2019 May.
Article in English | MEDLINE | ID: mdl-31007752

ABSTRACT

Previous studies by our group have indicated that exercise intervention can ameliorate endothelial dysfunction, which is an early pathophysiological change of prediabetes mellitus. The present study aimed to test the hypothesis that nitric oxide synthases (NOSs), which are expressed in blood vessel endothelium, contribute to the mitigation of vascular endothelium-dependent dysfunction by aerobic exercise in prediabetes mellitus. A prediabetic rat model was established by feeding the rats an additional high-energy diet, and was confirmed by testing blood glucose levels, the area-under-the-curve for the blood glucose tests (P<0.05) and the changes to the histological morphology of the thoracic aorta. Further examination identified that NOS expression changed significantly between the control and prediabetes groups, indicating endothelial dysfunction in the prediabetic rats. Following aerobic exercise, a significant increase in NOS, endothelial (eNOS) mRNA and protein expression (P<0.05), and a significant decrease in NOS, inducible (iNOS) mRNA and protein expression (P<0.05) was identified in the prediabetic rats compared with the control group. No significant change in nitric oxide synthase, brain expression was observed in the prediabetic rat group compared with the control group. Notably, there was also a significant increase and decrease in eNOS and iNOS activity, respectively, in the prediabetes group compared with the control group (P<0.05). Furthermore, nitric oxide (NO) concentration in the vascular endothelium was detected, which revealed a significant increase in NO concentration in the prediabetic rats following aerobic exercise compared with the control (P<0.05). The present study provided results that demonstrated that aerobic exercise ameliorated the vascular endothelium-dependent dysfunction through the NOS/NO signaling pathway, which is primarily regulated by NOS expression and activity, in prediabetes mellitus. The current study provided the theoretical basis for the use of exercise intervention to prevent diabetes mellitus during the early stage.

15.
Int J Biol Macromol ; 127: 311-319, 2019 Apr 15.
Article in English | MEDLINE | ID: mdl-30639594

ABSTRACT

Chitosan has been made into various hemostats, but their hemostatic efficiency for controlling severe traumatic bleeding is still inadequate. The aim of this work is to make quick hemostats by incorporating mesoporous silica nanoparticles into chitosan. Porous chitosan-silica composite microspheres (CSMS-S) with high hemostatic efficacy were fabricated through a combination of the microemulsion, thermally induced phase separation, and surfactant templating method. A large number of mesoporous silica nanoparticles were formed on and within the CSMS-S microspheres, which had abundant surface and inner macropores. The synergetic two hemostatic mechanisms from chitosan and mesoporous silica nanoparticles let CSMS-S composite microspheres with proper amount of silica displayed better hemostatic potential than the single component porous chitosan microspheres (CSMS). Within a same time interval, the whole blood clotting kinetics showed that CSMS-S could form larger blood clots than CSMS. The hemostatic time of CSMS-S was down to 97 s from 114 s of CSMS in the rat liver laceration model. The cytotoxicity and histological analysis proved that CSMS-S was a safe hemostatic agent without noticeable adverse effects on tissues around the wound. Our results demonstrate that CSMS-K is a promising quick hemostatic agent for traumatic hemorrhaging control.


Subject(s)
Chitosan , Hemorrhage , Microspheres , Nanoparticles , Silicon Dioxide , Wounds and Injuries , Animals , Chitosan/chemistry , Chitosan/pharmacology , Hemorrhage/blood , Hemorrhage/drug therapy , Hemorrhage/pathology , Mice , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Rats , Silicon Dioxide/chemistry , Silicon Dioxide/pharmacology , Wounds and Injuries/blood , Wounds and Injuries/drug therapy , Wounds and Injuries/pathology
16.
Cells ; 9(1)2019 Dec 27.
Article in English | MEDLINE | ID: mdl-31892155

ABSTRACT

The contribution of autophagy to catabolic balance has been well-established in various types of cells, whereas the involvement of autophagy in progesterone synthesis during rat pregnancy still remains unknown. Therefore, the present study was designed to evaluate the role of autophagy in progesterone production during the luteal development of pregnant rats. The results showed autophagy-related proteins was maintained at a low level on day 10 after pregnancy, significantly induced on day 16 and subsided to a relative low level on day 21, which was consistent with the changes of serum progesterone levels. The findings further indicated the contribution of autophagy to progesterone production was regulated by inactivation of Akt/mTOR signaling during the luteal development of pregnant rats in in vivo and in vitro experiments. Further investigations revealed autophagy may be involved in the surge of progesterone production in pregnant rats, as inhibition of autophagy by 3-MA compromised serum progesterone levels. Furthermore, 3-MA treatment also leveled down the number of lipid droplets in luteal cells, implying that autophagy may affect the production of progesterone by manipulating the formation of lipid droplets in luteal cells. In addition, the results suggested that mitophagy was mobilized during the primary stage of luteolysis and inhibition of autophagy promoted the increase of redundant mitochondrial and cytoplasmic cytochrome C in luteal cells of pregnant rats. Taken together, the present study indicated that autophagy-related proteins were induced by the inactivation of Akt/mTOR signaling and then contributed to the progesterone production possibly by affecting the formation of intracellular lipid droplets during the luteal development of pregnant rats. To our knowledge, this will provide a new insight into the important mechanism of autophagy regulating progesterone production in ovaries of pregnant mammals.


Subject(s)
Autophagy , Corpus Luteum/physiology , Progesterone/biosynthesis , Animals , Autophagy/drug effects , Autophagy/genetics , Biomarkers , Female , Gene Expression Regulation , Homeostasis , Immunohistochemistry , Lipid Metabolism/drug effects , Luteal Cells/metabolism , Male , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/metabolism , Pregnancy , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
17.
Mater Sci Eng C Mater Biol Appl ; 85: 27-36, 2018 Apr 01.
Article in English | MEDLINE | ID: mdl-29407154

ABSTRACT

Quick hemostats for non-lethal massive traumatic bleeding in battlefield and civilian accidents are important for reducing mortality and medical costs. Chitosan (CS) has been widely used as a clinic hemostat. To enhance its hemostatic efficiency, Zn2+ in the form of zinc alginate (ZnAlg) was introduced to CS to make porous CS@ZnAlg microspheres with ZnAlg component on the surface. Such microspheres were prepared by successive steps of micro-emulsion, polyelectrolyte adhesion, and thermally induced phase separation. Their structure and hemostatic performance were analyzed by SEM, FT-IR, XPS and a series of in vitro hemostatic experiments including thromboelastography analysis. The composite microspheres had an outer and internal interconnected porous structure. Their size, surface area, and water absorption ratio were ca. 70µm, 48m2/g, and 1850%, respectively. Compared to the neat chitosan microspheres, the CS@ZnAlg microspheres showed shorter onset of clot formation, much faster in vitro and in vivo whole blood clotting, bigger clot, less blood loss, and shorter hemostatic time in the rat liver laceration and tail amputation models. The synergetic hemostatic effects from (1) the electrostatic attraction between chitosan component and red blood cells, (2) the activation of coagulation factor XII by Zn2+ of zinc alginate component, and (3) physical blocking by microsphere matrix, contributed to the enhanced hemostatic performance of CS@ZnAlg microspheres.


Subject(s)
Chitosan/chemistry , Hemostasis , Microspheres , Thrombosis/therapy , Zinc/chemistry , Alginates/chemistry , Animals , Blood Coagulation , Cell Death , Elasticity Imaging Techniques , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Ions , Kinetics , Liver/pathology , Mice , Photoelectron Spectroscopy , Porosity , Rats, Sprague-Dawley , Spectroscopy, Fourier Transform Infrared , Tail , Thermogravimetry
18.
Exp Ther Med ; 14(6): 5981-5989, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29250140

ABSTRACT

Dimethyl carbonate (DMC) is a widely used industrial chemical, which may be increasingly used in the future. However, its toxicity profile remains largely unknown. The present study was designed to investigate the effects of DMC exposure on the ovaries and the effect of autophagy activation on follicular development. Rats were randomly divided into a control group and low, medium and high dose DMC groups (all n=10). Histological analyses identified no marked differences in the rate of apoptosis between the control and low dose groups; however, marked apoptosis occurred in the medium and high dose groups. The expression of cleaved caspase-3 was significantly increased in the medium and high dose groups, which was consistent with changes observed in the expression of Bcl-2 and Bax. These results indicated that DMC exposure induces toxicity on ovarian function via the induction of apoptosis. The increased expression of the autophagy-related proteins light chain 3II, beclin-1 and p62 following exposure to DMC further indicated that autophagy was activated primarily in the granulosa cells of ovarian follicles in a dose-dependent manner. In addition, the changes in the expression of hypoxia inducible factor 1 α subunit (HIF-1α) and its target protein BCL2 interacting protein 3 (BNIP3) indicated that they may serve a role in the follicular development process induced by DMC. The results of the current study demonstrated that DMC exposure activated autophagy in the ovarian tissue. Furthermore, exposure to low doses of DMC may protect follicular development by activating the HIF-1α/BNIP3 signaling pathway. Taken together, these results indicate that exposure to medium and high doses of DMC induced follicular atresia by activating the apoptotic signaling pathway. This may be an important mechanism of regulating follicular development and ovarian function in mammals.

19.
Carbohydr Polym ; 177: 135-143, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-28962752

ABSTRACT

The hemostatic performance of chitosan was greatly improved by blending it with kaolin to fabricate porous composite microspheres (CSMS-K) through inverse emulsion method combining with thermally induced phase separation. The CSMS-K had high amount of interior and surface pores. The synergetic hemostatic competence of chitosan and kaolin components made the hemostatic efficacy of CSMS-K superior to chitosan porous microspheres (CSMS). The hemostatic time of CSMS-K3 in the rat tail amputation and liver laceration models was down to respective 120 and 99s from 183 and 134s of CSMS, and the blood loss of CSMS-K3 was respectively 65% and 36% of that of CSMS in the rat tail amputation and liver laceration models. The whole blood clotting kinetics proved that CSMS-K3 formed larger blood clots than CSMS and Celox within a same time period. Our results suggested that the CSMS-K is a potential quick pro-coagulant agent for traumatic hemorrhaging control.


Subject(s)
Chitosan/chemistry , Chitosan/pharmacology , Kaolin/chemistry , Kaolin/pharmacology , Liver/drug effects , Microspheres , Tail/drug effects , Animals , Hemostatics/chemistry , Hemostatics/pharmacology , Liver/injuries , Porosity , Rats , Tail/injuries
20.
Mater Sci Eng C Mater Biol Appl ; 77: 411-419, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28532047

ABSTRACT

Controlling massive hemorrhage is of great importance to lower transfusional medical cost, and to reduce death and mobility rate in battlefield and civilian accidents. We reported the fabrication of porous chitosan microspheres (CSMS) with tunable surface pore size by microemulsion combined with thermally induced phase separation technique, and its application as a quick hemostat. Their hemostatic property was characterized by blood clotting kinetics, adherence interaction between red blood cells/platelets and CSMS, in vitro and in vivo hemostasis by rat tail amputation and liver laceration models, and histological analysis. Their density, surface area, porosity, water absorption ratio were 0.04-0.06g/cm3, 28.2-31.5m2/g, 98%, and 15.5-23.2g/g, respectively. The surface pore was controlled to be smaller than 2.0µm. The porous CSMS showed increasing hemostatic efficacy with decreasing surface pore size. Compared to the conventional compact chitosan particles (CCSP), the porous CSMS had much improved in vitro and in vivo hemostatic potential with respect to formation of blood clot, hemostatic time, and blood loss. For instance, the hemostatic time and blood loss of CSMS in the rat liver laceration model were down to respectively 70s and 0.026g from 175s and 0.28g of CCSP. Histological examination showed that application of porous CSMS to liver laceration caused no destruction of underlying hepatocytes, inflammatory reaction, and thermal injury to liver tissue. The porous CSMS is a biodegradable, quick and safe hemostat, which can be used in various wounds including complex and non-compressive ones.


Subject(s)
Microspheres , Animals , Chitosan , Hemorrhage , Hemostatics , Porosity , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...