Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Int J Mol Sci ; 23(10)2022 May 18.
Article in English | MEDLINE | ID: mdl-35628454

ABSTRACT

We have previously described that placental activation of autophagy is a central feature of normal pregnancy, whereas autophagy is impaired in preeclampsia (PE). Here, we show that hypoxia-reoxygenation (H/R) treatment dysregulates key molecules that maintain autophagy-lysosomal flux in primary human trophoblasts (PHTs). Ultrastructural analysis using transmission electron microscopy reveals a significant reduction in autophagosomes and autolysosomes in H/R-exposed PHTs. H/R-induced accumulation of protein aggregates follows a similar pattern that occurs in PHTs treated with a lysosomal disruptor, chloroquine. Importantly, the placenta from early-onset PE deliveries exhibits the same features as seen in H/R-treated PHTs. Taken together, our results indicate that H/R disrupts autophagic machinery in PHTs and that impaired autophagy in the placenta from early-onset PE deliveries mimics the events in H/R-treated PHTs. Notably, assessment of key regulators at each stage of autophagic processes, especially lysosomal integrity, and verification of autophagic ultrastructure are essential for an accurate evaluation of autophagy activity in human trophoblasts and placental tissue from PE deliveries.


Subject(s)
Pre-Eclampsia , Trophoblasts , Autophagy/physiology , Female , Humans , Hypoxia/metabolism , Lysosomes/metabolism , Placenta/metabolism , Pre-Eclampsia/metabolism , Pregnancy , Trophoblasts/metabolism
2.
Int J Mol Sci ; 22(5)2021 Feb 28.
Article in English | MEDLINE | ID: mdl-33670947

ABSTRACT

Aggrephagy is defined as the selective degradation of aggregated proteins by autophagosomes. Protein aggregation in organs and cells has been highlighted as a cause of multiple diseases, including neurodegenerative diseases, cardiac failure, and renal failure. Aggregates could pose a hazard for cell survival. Cells exhibit three main mechanisms against the accumulation of aggregates: protein refolding by upregulation of chaperones, reduction of protein overload by translational inhibition, and protein degradation by the ubiquitin-proteasome and autophagy-lysosome systems. Deletion of autophagy-related genes reportedly contributes to intracellular protein aggregation in vivo. Some proteins recognized in aggregates in preeclamptic placentas include those involved in neurodegenerative diseases. As aggregates are derived both intracellularly and extracellularly, special endocytosis for extracellular aggregates also employs the autophagy machinery. In this review, we discuss how the deficiency of aggrephagy and/or macroautophagy leads to poor placentation, resulting in preeclampsia or fetal growth restriction.


Subject(s)
Macroautophagy , Placenta/physiopathology , Pre-Eclampsia/physiopathology , Animals , Female , Humans , Lysosomes/metabolism , Placenta/metabolism , Pre-Eclampsia/etiology , Pre-Eclampsia/metabolism , Pregnancy , Protein Aggregation, Pathological
3.
Article in English | MEDLINE | ID: mdl-32939889

ABSTRACT

Hypertensive disorders of pregnancy, including preeclampsia, directly affect maternal and perinatal morbidity and mortality. As the pathophysiology of preeclampsia is multi-factorial and has been studied using different approaches, we have demonstrated that impaired autophagy is an intertwined risk factor for preeclampsia. This concept has been verified in both in vitro and in vivo experiments. Autophagy is primarily involved in maintaining cellular homeostasis, and in immune regulation, longevity, cytokines secretion and a variety of other biological functions. Here, we review the role of autophagy in normal embryogenesis and placentation. Once placental autophagy is impaired by metabolic stress such as hypoxia, endoplasmic reticulum stress or starvation, placental development could be disrupted, resulting in functional maladaptations at the maternal-fetal interface. These malfunctions may result in fetal growth restriction or preeclampsia.

4.
Int J Mol Sci ; 21(9)2020 May 07.
Article in English | MEDLINE | ID: mdl-32392703

ABSTRACT

Placental homeostasis is directly linked to fetal well-being and normal fetal growth. Placentas are sensitive to various environmental stressors, including hypoxia, endoplasmic reticulum stress, and oxidative stress. Once placental homeostasis is disrupted, the placenta may rebel against the mother and fetus. Autophagy is an evolutionally conservative mechanism for the maintenance of cellular and organic homeostasis. Evidence suggests that autophagy plays a crucial role throughout pregnancy, including fertilization, placentation, and delivery in human and mouse models. This study reviews the available literature discussing the role of autophagy in preeclampsia.


Subject(s)
Placenta/physiopathology , Pre-Eclampsia/physiopathology , Autophagy , Endoplasmic Reticulum Stress , Female , Homeostasis , Humans , Oxidative Stress , Placenta/metabolism , Pre-Eclampsia/metabolism , Pregnancy , Signal Transduction
5.
Int J Mol Sci ; 20(9)2019 May 11.
Article in English | MEDLINE | ID: mdl-31083536

ABSTRACT

Autophagy is an evolutionarily conserved process in eukaryotes to maintain cellular homeostasis under environmental stress. Intracellular control is exerted to produce energy or maintain intracellular protein quality controls. Autophagy plays an important role in embryogenesis, implantation, and maintenance of pregnancy. This role includes supporting extravillous trophoblasts (EVTs) that invade the decidua (endometrium) until the first third of uterine myometrium and migrate along the lumina of spiral arterioles under hypoxic and low-nutrient conditions in early pregnancy. In addition, autophagy inhibition has been linked to poor placentation-a feature of preeclamptic placentas-in a placenta-specific autophagy knockout mouse model. Studies of autophagy in human placentas have revealed controversial results, especially with regard to preeclampsia and gestational diabetes mellitus (GDM). Without precise estimation of autophagy flux, wrong interpretation would lead to fixed tissues. This paper presents a review of the role of autophagy in pregnancy and elaborates on the interpretation of autophagy in human placental tissues.


Subject(s)
Autophagy , Animals , Autophagy/genetics , Female , Humans , Models, Biological , Placentation , Pregnancy , Pregnancy Complications/pathology , Reproduction
6.
J Obstet Gynaecol Res ; 43(4): 633-643, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28418212

ABSTRACT

Autophagy is a well-conserved mechanism in cells from yeast to mammals, and autophagy maintains homeostasis against stress. The role of autophagy was originally shown to be a mechanism of energy production under starvation. In fact, multiple lines of evidence reveal that autophagy has numerous functions, such as protection from stress, energy regulation, immune regulation, differentiation, proliferation, and cell death. In the field of reproduction, the role of autophagy in implantation, embryogenesis, placentation, and delivery has become clearer. In addition, recent study has elucidated that the placenta has the ability to protect extraplacental cells from virus infection by activating autophagy. During resent research into autophagy, several issues have occurred in the interpretation of the autophagy status. In this review, we discuss the relation between autophagy and reproductive events, and show the importance of autophagy for placentation and pre-eclampsia.


Subject(s)
Autophagy/physiology , Embryo Implantation/physiology , Embryonic Development/physiology , Oogenesis/physiology , Placentation/physiology , Pre-Eclampsia/physiopathology , Animals , Female , Humans , Pregnancy
7.
J Am Soc Nephrol ; 26(11): 2716-29, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25788532

ABSTRACT

Hyperuricemia is an independent risk factor for CKD and contributes to kidney fibrosis. In this study, we investigated the effect of EGF receptor (EGFR) inhibition on the development of hyperuricemic nephropathy (HN) and the mechanisms involved. In a rat model of HN induced by feeding a mixture of adenine and potassium oxonate, increased EGFR phosphorylation and severe glomerular sclerosis and renal interstitial fibrosis were evident, accompanied by renal dysfunction and increased urine microalbumin excretion. Administration of gefitinib, a highly selective EGFR inhibitor, prevented renal dysfunction, reduced urine microalbumin, and inhibited activation of renal interstitial fibroblasts and expression of extracellular proteins. Gefitinib treatment also inhibited hyperuricemia-induced activation of the TGF-ß1 and NF-κB signaling pathways and expression of multiple profibrogenic cytokines/chemokines in the kidney. Furthermore, gefitinib treatment suppressed xanthine oxidase activity, which mediates uric acid production, and preserved expression of organic anion transporters 1 and 3, which promotes uric acid excretion in the kidney of hyperuricemic rats. Thus, blocking EGFR can attenuate development of HN via suppression of TGF-ß1 signaling and inflammation and promotion of the molecular processes that reduce uric acid accumulation in the body.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Hyperuricemia/drug therapy , Hyperuricemia/metabolism , Kidney Diseases/metabolism , Animals , Chemokines/metabolism , Cytokines/metabolism , Disease Progression , ErbB Receptors/metabolism , Fibroblasts/metabolism , Fibrosis/pathology , Gefitinib , Inflammation , Kidney/metabolism , Kidney/pathology , Male , Phosphorylation , Quinazolines/therapeutic use , Rats , Rats, Sprague-Dawley , Risk Factors , Signal Transduction , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1/metabolism , Uric Acid/chemistry
8.
Am J Pathol ; 183(5): 1425-1436, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24035612

ABSTRACT

Preeclampsia is a major pregnancy complication with potential short- and long-term consequences for both mother and fetus. Understanding its pathogenesis and causative biomarkers is likely to yield insights for prediction and treatment. Herein, we provide evidence that transthyretin, a transporter of thyroxine and retinol, is aggregated in preeclampsia and is present at reduced levels in sera of preeclamptic women, as detected by proteomic screen. We demonstrate that transthyretin aggregates form deposits in preeclampsia placental tissue and cause apoptosis. By using in vitro approaches and a humanized mouse model, we provide evidence for a causal link between dysregulated transthyretin and preeclampsia. Native transthyretin inhibits all preeclampsia-like features in the humanized mouse model, including new-onset proteinuria, increased blood pressure, glomerular endotheliosis, and production of anti-angiogenic factors. Our findings suggest that a focus on transthyretin structure and function is a novel strategy to understand and combat preeclampsia.


Subject(s)
Pre-Eclampsia/metabolism , Pre-Eclampsia/pathology , Prealbumin/metabolism , Animals , Disease Models, Animal , Endoglin , Female , Humans , Immunoprecipitation , Interleukin-10/deficiency , Interleukin-10/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Neovascularization, Physiologic , Pre-Eclampsia/blood , Prealbumin/chemistry , Pregnancy , Protein Binding , Protein Structure, Quaternary , Proteomics , Solubility , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Vascular Endothelial Growth Factor Receptor-1/metabolism
9.
Exp Mol Med ; 56(5): 1206-1220, 2024 May.
Article in English | MEDLINE | ID: mdl-38760513

ABSTRACT

The etiology of preeclampsia (PE), a severe complication of pregnancy with several clinical manifestations and a high incidence of maternal and fetal morbidity and mortality, remains unclear. This issue is a major hurdle for effective treatment strategies. We recently demonstrated that PE exhibits an Alzheimer-like etiology of impaired autophagy and proteinopathy in the placenta. Targeting of these pathological pathways may be a novel therapeutic strategy for PE. Stimulation of autophagy with the natural disaccharide trehalose and its lacto analog lactotrehalose in hypoxia-exposed primary human trophoblasts restored autophagy, inhibited the accumulation of toxic protein aggregates, and restored the ultrastructural features of autophagosomes and autolysosomes. Importantly, trehalose and lactotrehalose inhibited the onset of PE-like features in a humanized mouse model by normalizing autophagy and inhibiting protein aggregation in the placenta. These disaccharides restored the autophagy-lysosomal biogenesis machinery by increasing nuclear translocation of the master transcriptional regulator TFEB. RNA-seq analysis of the placentas of mice with PE indicated the normalization of the PE-associated transcriptome profile in response to trehalose and lactotrehalose. In summary, our results provide a novel molecular rationale for impaired autophagy and proteinopathy in patients with PE and identify treatment with trehalose and its lacto analog as promising therapeutic options for this severe pregnancy complication.


Subject(s)
Autophagy , Lysosomes , Pre-Eclampsia , Trehalose , Autophagy/drug effects , Pre-Eclampsia/drug therapy , Pre-Eclampsia/metabolism , Female , Humans , Pregnancy , Animals , Lysosomes/metabolism , Lysosomes/drug effects , Trehalose/analogs & derivatives , Trehalose/pharmacology , Trehalose/therapeutic use , Mice , Trophoblasts/metabolism , Trophoblasts/drug effects , Trophoblasts/pathology , Placenta/metabolism , Placenta/drug effects , Disease Models, Animal
10.
J Biol Chem ; 287(17): 14178-91, 2012 Apr 20.
Article in English | MEDLINE | ID: mdl-22378779

ABSTRACT

Receptor down-modulation is the key mechanism by which G protein-coupled receptors (GPCRs) prevent excessive receptor signaling in response to agonist stimulation. Recently, the trans-Golgi network (TGN) has been implicated as a key checkpoint for receptor endocytosis and degradation. Here, we investigated the involvement of the TGN in down-modulation of ß1-adrenergic receptor in response to persistent isoprotenerol stimulation. Immunofluorescent staining showed that ~50% of endocytosed ß1AR colocalized with TGN-46 at 5 h. Disruption of the TGN by brefeldin A (BFA) led to the robust accumulation of endocytosed ß1AR in Rab11(+) recycling endosomes, inhibited ß1AR entry into LAMP1(+) lysosomes, and as a result enhanced ß1AR recycling to the plasma membrane. The lysosomotropic agent, chloroquine, arrested the majority of endocytosed ß1AR in the TGN by 4 h. Immunoblot analysis showed that either disruption of the TGN or blockage of the lysosome prevented ß1AR degradation. Co-expression of GFP-arrestin-3 in ß1AR cells increased the endocytosis of ß1AR and facilitated its entry to the TGN but inhibited recycling to the plasma membrane. Arrestin-3-induced inhibition of ß1AR recycling was reversed by BFA treatment, whereas chloroquine induced the accumulation of arrestin-3 with ß1AR in the TGN. These results demonstrate for the first time that the TGN acts as a checkpoint for both the recycling and down-regulation of ß1AR and that arrestin-3 not only mediates ß1AR endocytosis but also its recycling through the TGN.


Subject(s)
Receptors, Adrenergic, beta-1/metabolism , trans-Golgi Network/metabolism , Arrestins/metabolism , Brefeldin A/pharmacology , Cell Membrane/metabolism , Down-Regulation , Endocytosis , Endosomes/metabolism , Golgi Apparatus/metabolism , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Lysosomes/metabolism , Models, Biological , Proteasome Endopeptidase Complex/metabolism
11.
Biology (Basel) ; 12(8)2023 Jul 26.
Article in English | MEDLINE | ID: mdl-37626934

ABSTRACT

Our prior studies have shown that protein misfolding and aggregation in the placenta are linked to the development of preeclampsia, a severe pregnancy complication. We identified transthyretin (TTR) as a key component of the aggregated protein complex. However, the regulation of native TTR in normal pregnancy remains unclear. In this study, we found that pregnant mice exhibited a remarkable and progressive decline in serum TTR levels through gestational day (gd) 12-14, followed by an increase in late pregnancy and postpartum. Meanwhile, serum albumin levels showed a modest but statistically significant increase throughout gestation. TTR protein and mRNA levels in the liver, a primary source of circulating TTR, mirrored the changes observed in serum TTR levels during gestation. Intriguingly, a similar pattern of TTR alteration was also observed in the serum of pregnant women and pregnant interleukin-10-knockout (IL-10-/-) mice with high inflammation background. In non-pregnant IL-10-/- mice, serum TTR levels were significantly lower than those in age-matched wild-type mice. Administration of IL-10 to non-pregnant IL-10-/- mice restored their serum TTR levels. Notably, dysregulation of TTR resulted in fewer implantation units, lower fetal weight, and smaller litter sizes in human TTR-overexpressing transgenic mice. Thus, TTR may play a pivotal role as a crucial regulator in normal pregnancy, and inflammation during pregnancy may contribute to the downregulation of serum TTR presence.

12.
Nat Commun ; 14(1): 5414, 2023 09 05.
Article in English | MEDLINE | ID: mdl-37669931

ABSTRACT

Preeclampsia (PE) is the leading cause of maternal and fetal mortality globally and may trigger dementia later in life in mothers and their offspring. However, the etiological drivers remain elusive. Cis P-tau is an early etiological driver and blood biomarker in pre-clinical Alzheimer's and after vascular or traumatic brain injury, which can be targeted by stereo-specific antibody, with clinical trials ongoing. Here we find significant cis P-tau in the placenta and serum of PE patients, and in primary human trophoblasts exposed to hypoxia or sera from PE patients due to Pin1 inactivation. Depletion of cis P-tau from PE patient sera by the antibody prevents their ability to disrupt trophoblast invasion and endovascular activity and to cause the PE-like pathological and clinical features in pregnant humanized tau mice. Our studies uncover that cis P-tau is a central circulating etiological driver and its stereo-specific antibody is valuable for early PE diagnosis and treatment.


Subject(s)
Placenta , Pre-Eclampsia , Female , Pregnancy , Humans , Animals , Mice , Causality , Trophoblasts , Antibodies , Mothers
13.
J Reprod Immunol ; 155: 103781, 2023 02.
Article in English | MEDLINE | ID: mdl-36463798

ABSTRACT

Autophagy is a lysosomal degradation pathway that supports metabolic adaptation and energy cycling. It is essential for cell homeostasis, differentiation, development, and survival. Recent studies have shown that autophagy could influence immune responses by regulating immune cell functions. Reciprocally, immune cells strongly influence autophagy. Immune cells at the maternal-fetal interface are thought to play essential roles in pregnancy. Here, we review the induction of autophagy at the maternal-fetal interface and its role in decidualization and placental development. Additionally, we emphasize the role of autophagy in the immune microenvironment at the maternal-fetal interface, including innate immunity, adaptive immunity, and immune tolerance molecules. It also suggests new research directions and prospects.


Subject(s)
Immunity, Innate , Placenta , Humans , Pregnancy , Female , Adaptive Immunity , Immune Tolerance , Autophagy , Maternal-Fetal Exchange
14.
Biology (Basel) ; 12(3)2023 Feb 26.
Article in English | MEDLINE | ID: mdl-36979065

ABSTRACT

Autophagy is a fundamental process involved in regulating cellular homeostasis. Autophagy has been classically discovered as a cellular process that degrades cytoplasmic components non-selectively to produce energy. Over the past few decades, this process has been shown to work in energy production, as well as in the reduction of excessive proteins, damaged organelles, and membrane trafficking. It contributes to many human diseases, such as neurodegenerative diseases, carcinogenesis, diabetes mellitus, development, longevity, and reproduction. In this review, we provide important information for interpreting results related to autophagic experiments and present the role of autophagy in this field.

15.
J Reprod Immunol ; 155: 103766, 2023 02.
Article in English | MEDLINE | ID: mdl-36470134

ABSTRACT

Inhibition of autophagy contributes to the pathophysiology of preeclampsia. Although chloroquine (CHQ) is an autophagy inhibitor, it can reduce the occurrence of preeclampsia in women with systemic lupus erythematosus. To clarify this important clinical question, this study aimed to address the safety of CHQ in trophoblast cells from the viewpoint of homeostasis, in which the anti-oxidative stress (OS) response and autophagy are involved. We used Western blotting to evaluate the protein levels in the trophoblast cells. The expression levels of heme oxygenase-1 (HO-1), an anti-OS enzyme, mediate resistance to OS induced by hydrogen peroxide (H2O2) in trophoblast cell lines. Among the autophagy modulators, bafilomycin A1 (BAF), an autophagy inhibitor, but not autophagy activators, suppressed HO-1 expression in BeWo cells; CHQ did not suppress HO-1 expression in BeWo cells. To clarify the role of autophagy in HO-1 induction, we observed no difference in HO-1 induction by H2O2 between autophagy-normal and autophagy-deficient cells. As for the mechanism of HO-1 induction by OS, BAF suppressed HO-1 induction by downregulating the expression of neighbor of BRCA1 gene 1 (NBR1) in the selective p62-NBR1-nuclear factor erythroid 2-related factor 2 (Nrf2) autophagy pathway. CHQ did not inhibit HO-1 expression by sustaining NBR1 expression in human villous tissues compared to BAF treatment. In conclusion, CHQ is a safer medicine than BAF for sustaining NBR1, which resist against OS in trophoblasts by connecting selective autophagy and the anti-OS response.


Subject(s)
Antioxidants , Pre-Eclampsia , Pregnancy , Humans , Female , Antioxidants/metabolism , Antioxidants/pharmacology , Trophoblasts/metabolism , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Chloroquine/pharmacology , Chloroquine/metabolism , Pre-Eclampsia/drug therapy , Pre-Eclampsia/metabolism , Signal Transduction , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism
16.
J Biol Chem ; 286(25): 22441-55, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21540189

ABSTRACT

GPER is a G(s)-coupled seven-transmembrane receptor that has been linked to specific estrogen binding and signaling activities that are manifested by plasma membrane-associated enzymes. However, in many cell types, GPER is predominately localized to the endoplasmic reticulum (ER), and only minor amounts of receptor are detectable at the cell surface, an observation that has caused controversy regarding its role as a plasma membrane estrogen receptor. Here, we show that GPER constitutively buds intracellularly into EEA-1+ endosomes from clathrin-coated pits. Nonvisual arrestins-2/-3 do not co-localize with GPER, and expression of arrestin-2 dominant-negative mutants lacking clathrin- or ß-adaptin interaction sites fails to block GPER internalization suggesting that arrestins are not involved in GPER endocytosis. Like ß1AR, which recycles to the plasma membrane, GPER co-traffics with transferrin+, Rab11+ recycling endosomes. However, endocytosed GPER does not recycle to the cell surface, but instead returns to the trans-Golgi network (TGN) and does not re-enter the ER. GPER is ubiquitinated at the cell surface, exhibits a short half-life (t½;) <1 h), and is protected from degradation by the proteasome inhibitor, MG132. Disruption of the TGN by brefeldin A induces the accumulation of endocytosed GPER in Rab11+ perinuclear endosomes and prevents GPER degradation. Our results provide an explanation as to why GPER is not readily detected on the cell surface in some cell types and further suggest that TGN serves as the checkpoint for degradation of endocytosed GPER.


Subject(s)
Down-Regulation , Endocytosis , Proteasome Endopeptidase Complex/metabolism , Receptors, G-Protein-Coupled/metabolism , trans-Golgi Network/metabolism , Animals , Cattle , Cell Membrane/metabolism , Clathrin/metabolism , Endosomes/metabolism , HEK293 Cells , Humans , Kinetics , Mice , Receptors, Adrenergic, beta-1/metabolism , Receptors, CXCR4/metabolism , Recombinant Fusion Proteins/metabolism , Ubiquitin/metabolism
17.
Hypertension ; 79(8): 1738-1754, 2022 08.
Article in English | MEDLINE | ID: mdl-35607996

ABSTRACT

BACKGROUND: We have demonstrated that protein aggregation plays a pivotal role in the pathophysiology of preeclampsia and identified several aggregated proteins in the circulation of preeclampsia patients, the most prominent of which is the serum protein TTR (transthyretin). However, the mechanisms that underlie protein aggregation remain poorly addressed. METHODS: We examined TTR aggregates in hypoxia/reoxygenation-exposed primary human trophoblasts (PHTs) and the preeclampsia placenta using complementary approaches, including a novel protein aggregate detection assay. Mechanistic analysis was performed in hypoxia/reoxygenation-exposed PHTs and Ttr transgenic mice overexpressing transgene-encoded wild-type human TTR or Ttr-/- mice. High-resolution ultrasound analysis was used to measure placental blood flow in pregnant mice. RESULTS: TTR aggregation was inducible in PHTs and the TCL-1 trophoblast cell line by endoplasmic reticulum stress inducers or autophagy-lysosomal disruptors. PHTs exposed to hypoxia/reoxygenation showed increased intracellular BiP (binding immunoglobulin protein), phosphorylated IRE1α (inositol-requiring enzyme-1α), PDI (protein disulfide isomerase), and Ero-1, all markers of the unfolded protein response, and the apoptosis mediator caspase-3. Blockade of IRE1α inhibited hypoxia/reoxygenation-induced upregulation of Ero-1 in PHTs. Excessive unfolded protein response activation was observed in the early-onset preeclampsia placenta. Importantly, pregnant human TTR mice displayed aggregated TTR in the junctional zone of the placenta and severe preeclampsia-like features. High-resolution ultrasound analysis revealed low blood flow in uterine and umbilical arteries in human TTR mice compared with control mice. However, Ttr-/- mice did not show any pregnancy-associated abnormalities. CONCLUSIONS: These observations in the preeclampsia placenta, cultured trophoblasts, and Ttr transgenic mice indicate that TTR aggregation is an important causal contributor to preeclampsia pathophysiology.


Subject(s)
Pre-Eclampsia , Trophoblasts , Animals , Carrier Proteins/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , Endoribonucleases/metabolism , Female , Humans , Hypoxia/metabolism , Mice , Mice, Transgenic , Placenta/metabolism , Pre-Eclampsia/genetics , Pre-Eclampsia/metabolism , Prealbumin/analysis , Prealbumin/genetics , Prealbumin/metabolism , Pregnancy , Protein Aggregates , Protein Serine-Threonine Kinases , Trophoblasts/metabolism
18.
Int J Biol Sci ; 18(2): 599-616, 2022.
Article in English | MEDLINE | ID: mdl-35002512

ABSTRACT

Lactic acid (LA) metabolism in the tumor microenvironment contributes to the establishment and maintenance of immune tolerance. This pathway is characterized in tumor associated macrophages. However, the role and pathway of LA metabolism at maternal-fetal interface during early pregnancy, especially in decidual macrophage differentiation, are still unclear. Herein, for the first time, we discovered that LA can trigger either M2 or M1 macrophage polarization via oxidative phosphorylation and glycolysis regulation under normoxia or hypoxia, respectively. Also, LA metabolism played a vital role in decidual macrophages-mediated recurrent pregnancy loss (RPL), through HIF-1α/SRC/LDHA pathway. Moreover, blockade of LA intake with AZD3965 (MCT-1 inhibitor) could rescue pregnancy in an abortion-prone mouse model, suggesting a potential therapeutic target in RPL. Collectively, the present study identifies the previously unknown functions of LA metabolism in the differentiation of decidual macrophages in early normal pregnancy and RPL, and provides a potential therapeutic strategy in RPL by manipulating decidual macrophages' functions through LA metabolic pathway.


Subject(s)
Abortion, Spontaneous/metabolism , Lactic Acid/metabolism , Macrophages/metabolism , Pregnancy/metabolism , Trophoblasts/metabolism , Adult , Animals , Cell Differentiation , Disease Models, Animal , Female , Humans , L-Lactate Dehydrogenase/metabolism , Male , Maternal-Fetal Exchange , Mice , Mice, Inbred BALB C , Mice, Inbred CBA , Mice, Inbred DBA , Signal Transduction , src-Family Kinases/metabolism
19.
Am J Reprod Immunol ; 85(2): e13297, 2021 02.
Article in English | MEDLINE | ID: mdl-32619308

ABSTRACT

Pre-eclampsia is a hypertensive disease of pregnancy characterized by new-onset hypertension, with either proteinuria and/or organ dysfunction. Pre-eclampsia is a leading cause of maternal morbidity and mortality; however, the underlying cellular and molecular mechanisms are not well understood. There is consensus that the underlying mechanism(s) resulting in pre-eclampsia is centered around abnormal placentation, inadequate spiral-artery remodeling, and deficiency in trophoblast invasion, resulting in impaired maternal blood flow to the placenta and a release of signals and/or inflammatory mediators into maternal circulation triggering the systemic manifestations of pre-eclampsia. ER stress, resulting in impaired autophagy and placental release of aggregated proteins, may also confer systemic stress to maternal organs in pre-eclampsia. Extracellular vesicles (EVs), lipid-bilayer enclosed structures containing macromolecules including proteins, miRNA, and other important nucleotides, have been suggested to play an important role in this maternal-fetal communication. Circulating EVs are present in greater quantity in the plasma of pre-eclampsia subjects compared to normal pregnancy, and the placental derived EVs have been shown to have altered protein and RNA cargo. In this review, we will focus on EVs and their role in pre-eclampsia, specifically their role in immune responses, inflammation, altered angiogenesis, and endothelial dysfunction.


Subject(s)
Extracellular Vesicles/metabolism , Immunity/immunology , Inflammation/immunology , Placenta/metabolism , Pre-Eclampsia/immunology , Animals , Female , Humans , Neovascularization, Pathologic , Pregnancy
20.
Front Cell Infect Microbiol ; 11: 694298, 2021.
Article in English | MEDLINE | ID: mdl-34485175

ABSTRACT

Understanding of sterile inflammation and its associated biological triggers and diseases is still at the elementary stage. This becomes more warranted in cases where infections are not associated with the pathology. Detrimental effects of bacterial and viral infections on the immune responses at the maternal-fetal interface as well as pregnancy outcomes have been well documented. However, an infection-induced etiology is not thought to be a major contributing component to severe pregnancy complications such as preeclampsia (PE) and gestational diabetes. How is then an inflammatory signal thought to be associated with these pregnancy complications? It is not clear what type of inflammation is involved in the onset of PE-like features. We opine that sterile inflammation regulated by the inflammasome-gasdermins-caspase-1 axis is a contributory factor to the onset of PE. We hypothesize that increased production and release of damage-associated molecular patterns (DAMPs) or Alarmins such as high-mobility group box1 (HMGB1), cell-free fetal DNA, uric acid, the NOD-like receptor pyrin-containing receptor 3 (NLRP3) inflammasome, IL-1ß and IL-18 occur in the PE placenta. Some of these molecules have already been observed in the placenta from women with PE. Mechanistically, emerging evidence has demonstrated that excessive placental endoplasmic reticulum (ER) stress, impaired autophagy and gasdermine D (GSDMD)-mediated intrinsic pyroptosis are key events that contribute to systemic sterile inflammation in patients with PE, especially early-onset PE (e-PE). In this review, we highlight the advances on the roles of sterile inflammation and inflammatory signaling cascades involving ER stress, autophagy deficiency and pyroptosis in PE pathophysiology. Deciphering the mechanisms underlying these inflammatory pathways may provide potential diagnostic biomarkers and facilitate the development of therapeutic strategies to treat this devastating disease.


Subject(s)
Pre-Eclampsia , Female , Humans , Inflammasomes , Inflammation , NLR Family, Pyrin Domain-Containing 3 Protein , Placenta , Pre-Eclampsia/etiology , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL