Qian-Kun Lv’s research while affiliated with Soochow University and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (6)


New perspectives on the glymphatic system and the relationship between glymphatic system and neurodegenerative diseases
  • Literature Review

January 2025

·

27 Reads

·

1 Citation

Neurobiology of Disease

Yan-rui Sun

·

Qian-Kun Lv

·

Jun-Yi Liu

·

[...]

·


MT1 deletion impaired the phagocytic function of microglia. (A–C) Representative immunofluorescence images (A) and quantification (B, C) of fluorescence latex beads engulfed by WT and MT1‐KO primary microglial cells. Scale bar: 20.0 μm. Phagocytic cells (B) = number of cells phagocytosing latex beads/total cell count. Phagocytic capacity (C) = number of latex beads phagocytosed per single cell (n > 100 cells per group for analysis. T‐test, Mean ± SEM. *p < 0.05, ***p < 0.001 vs. WT group). (D–F) Representative immunofluorescence images (D) of fluorescence zymosan particles engulfed by WT and MT1‐KO primary microglial cells and the quantification of phagocytic cells (E) and phagocytic capacity (F). Scale bar: 20.0 μm (n > 100 cells per group for analysis. T‐test, Mean ± SEM. **p < 0.01, ***p < 0.001 vs. WT group).
MT1 deficiency impaired the phagocytosis and clearance of α‐Syn in microglia. (A) Immunofluorescence analyses reveal the phagocytic uptake of α‐Syn–GFP monomers by primary microglial cells derived from WT and MT1‐KO mice. Scale bar: 10.0 μm. (B) Transmission electron microscopy images depict the fibrous structure of both pre‐ and post‐sonication of α‐Syn PFF. Scale bar: 100.0 nm. (C, D) Western blot (C) and group data (D) for MT1 levels in primary microglial cells treated by α‐Syn PFF for 24 hours (n = 6. T‐test, Mean ± SEM. ***p < 0.001 vs. PBS group). (E–G) Western blot (E) and the quantification of α‐Syn monomer (F) and α‐Syn aggregates (G) in primary microglial cells from both WT and KO groups after a 6‐h exposure to PBS or α‐Syn PFF (10 μg/mL), followed by a medium change and a subsequent 24‐h incubation (n = 6. One‐way ANOVA followed by Tukey's post hoc analysis. Mean ± SEM. *p < 0.05, **p < 0.01 vs. as indicated).
MT1 regulated LC3‐associated phagocytosis in microglial cells. (A–D) The mRNA expression levels of Mtnr1a (A), Rubicon (B), Nox2 (C), and Uvrag (D) in BV2 cells 36 h post‐treatment with either negative control (NC) or si‐Mtnr1a small interfering RNA. 18S serving as internal controls (n = 3. T‐test, Mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001 vs. NC group). (E, F) Western blot image (E) of the protein expression level of MT1, Rubicon, PI3KC‐III, Beclin‐1, NOX2, ATG5, LC3B‐II in NC, and si‐Mtnr1a groups and their relative protein quantification (F). GAPDH serves as internal control (n = 3. T‐test, Mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001 vs. NC group). (G–K) Western blot image (G) of the protein expression level of Rubicon, PI3KC‐III, Beclin‐1, LC3B‐II in WT, and MT1‐KO microglia and their relative protein quantification (H–K). GAPDH serves as internal control (n = 3. T‐test, Mean ± SEM. *p < 0.05, **p < 0.01 vs. WT group).
MT1–Rubicon interaction: Enhancing LAP through MT1 overexpression or activation. (A) Co‐IP validation of MT1 and Rubicon interaction in MT1‐overexpressing BV2 cell line. (B–G) Western blot image (B) of the protein levels of MT1 and LAP‐related protein in BV2 cells transfecting Vector or MT1 overexpression plasmid for 36 h. The relative protein quantification of MT1 (C), LC3B‐II (D), Rubicon (E), NOX2 (F), and PI3KC‐III (G). GAPDH was used as internal controls (n = 3. T‐test, Mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001 vs. NC group). (H–N) Western blot image (H) of the protein levels of MT1 and LAP‐related protein in primary microglial cells transfecting NC or MT1 overexpression lentiviral for 120 h. The relative protein quantification of MT1 (I), Rubicon (J), PI3KC‐III (K), NOX2 (L), Beclin‐1 (M), and LC3B‐II1 (N). GAPDH was used as an internal control (n = 4. T‐test, Mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001 vs. NC group).
Microglial MT1 regulated the clearance and degradation of pathological α‐Syn in neurons. (A) Schematic depiction of the construction of the neuron–microglia co‐culture model. (B, C) Immunofluorescent image (B) of pS129 α‐Syn in co‐culture models of WT or MT1‐KO primary microglial cells with α‐Syn PFF‐induced pathological neurons, along with the corresponding statistical analysis of the number of pS129 α‐Syn (C) (MG = primary microglia, n > 10 cells per group for analysis. One‐way ANOVA followed by Tukey's post hoc analysis. Mean ± SEM. ***p < 0.001 vs. as indicated; ns, not significant). (D–I) Western blot images of the expression of α‐Syn in Triton‐insoluble (D) and Triton‐soluble (E) protein fractions of neuron–microglia co‐cultures. Quantitative analysis of Triton‐insoluble α‐Syn monomer (F) and aggregates (G), as well as Triton‐soluble α‐Syn monomer (H) and aggregates (I). GAPDH was used as control. (MG = primary microglia, n = 3. One‐way ANOVA followed by Tukey's post hoc analysis. Mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001 vs. as indicated; ns, not significant).
Microglial Melatonin Receptor 1 Degrades Pathological Alpha‐Synuclein Through Activating LC3‐Associated Phagocytosis In Vitro
  • Article
  • Full-text available

October 2024

·

32 Reads

·

1 Citation

Aims Parkinson's disease (PD) is characterized by the formation of Lewy bodies (LBs), primarily constituted of α‐synuclein (α‐Syn). Microglial cells exhibit specific reactivity toward misfolded proteins such as α‐Syn. However, the exact clearance mechanism and related molecular targets remain elusive. Methods BV2 cells, primary microglia from wild‐type and MT1 knockout mice, and primary cortical neurons were utilized as experimental models. The study investigated relevant mechanisms by modulating microglial MT1 expression through small RNA interference (RNAi) and lentiviral overexpression techniques. Furthermore, pathological aggregation of α‐Syn was induced using pre‐formed fibrils (PFF) α‐Syn. Co‐immunoprecipitation, immunofluorescence, Western blot (WB), and quantitative real‐time PCR were used to elucidate the mechanisms of molecular regulation. Results In this study, we elucidated the regulatory role of the melatonin receptor 1 (MT1) in the microglial phagocytic process. Following MT1 knockout, the ability of microglial cells to engulf latex beads and zymosan particles decreased, subsequently affecting the phagocytic degradation of fibrillar α‐Syn by microglial cells. Furthermore, the loss of MT1 receptors in microglial cells exacerbates the aggregation of α‐Syn in neurons induced by pre‐formed fibrils (PFF) α‐Syn. Mechanistically, MT1 influences the phagocytic function of microglial cells by regulating the Rubicon‐dependent LC3‐associated phagocytosis (LAP) pathway. Conclusion Taken together, the results suggest the neuroprotective function of microglial cells in clearing α‐Syn through MT1‐mediated LAP, highlighting the potential key role of MT1 in pathogenic mechanisms associated with α‐Syn.

Download

Melatonin MT1 receptors regulate the Sirt1/Nrf2/Ho-1/Gpx4 pathway to prevent α-synuclein-induced ferroptosis in Parkinson's disease

March 2024

·

15 Reads

·

23 Citations

Journal of Pineal Research

Parkinson's disease (PD) is a neurodegenerative disorder characterized by the loss of dopaminergic (DA) neurons and aggregation of α‐synuclein (α‐syn). Ferroptosis, a form of cell death induced by iron accumulation and lipid peroxidation, is involved in the pathogenesis of PD. It is unknown whether melatonin receptor 1 (MT1) modulates α‐syn and ferroptosis in PD. Here, we used α‐syn preformed fibrils (PFFs) to induce PD models in vivo and in vitro. In PD mice, α‐syn aggregation led to increased iron deposition and ferroptosis. MT1 knockout exacerbated these changes and resulted in more DA neuronal loss and severe motor impairment. MT1 knockout also suppressed the Sirt1/Nrf2/Ho1/Gpx4 pathway, reducing resistance to ferroptosis, and inhibited expression of ferritin Fth1, leading to more release of ferrous ions. In vitro experiments confirmed these findings. Knockdown of MT1 enhanced α‐syn PFF‐induced intracellular α‐syn aggregation and suppressed expression of the Sirt1/Nrf2/Ho1/Gpx4 pathway and Fth1 protein, thereby aggravating ferroptosis. Conversely, overexpression of MT1 reversed these effects. Our findings reveal a novel mechanism by which MT1 activation prevents α‐syn‐induced ferroptosis in PD, highlighting the neuroprotective role of MT1 in PD.


Fig. 1 Key neuroanatomical pathways of the circadian system. (1) SCN indirectly regulates melatonin release from the pineal gland by projecting light signals to PVN. (2) DMH receives light signals from SCN and then projects them to LC and VLPO, which in turn regulates sleep/awake activity. (3) SCN regulates thermoregulation and aggressive tendency by DMH through SPZ or not through SPZ. DMH dorsomedial hypothalamic nucleus; SPZ subparaventricular zone;
Circadian disruption and sleep disorders in neurodegeneration

February 2023

·

328 Reads

·

63 Citations

Translational Neurodegeneration

Disruptions of circadian rhythms and sleep cycles are common among neurodegenerative diseases and can occur at multiple levels. Accumulating evidence reveals a bidirectional relationship between disruptions of circadian rhythms and sleep cycles and neurodegenerative diseases. Circadian disruption and sleep disorders aggravate neurodegeneration and neurodegenerative diseases can in turn disrupt circadian rhythms and sleep. Importantly, circadian disruption and various sleep disorders can increase the risk of neurodegenerative diseases. Thus, harnessing the circadian biology findings from preclinical and translational research in neurodegenerative diseases is of importance for reducing risk of neurodegeneration and improving symptoms and quality of life of individuals with neurodegenerative disorders via approaches that normalize circadian in the context of precision medicine. In this review, we discuss the implications of circadian disruption and sleep disorders in neurodegenerative diseases by summarizing evidence from both human and animal studies, focusing on the bidirectional links of sleep and circadian rhythms with prevalent forms of neurodegeneration. These findings provide valuable insights into the pathogenesis of neurodegenerative diseases and suggest a promising role of circadian-based interventions.


α-Syn aggregates into two forms of oligomers or fibrils in the brain of PD patients, and finally forms LBs, which eventually lead to neuronal degeneration and loss. Created with BioRender.com
Neuroinflammation induced by aggregated α-syn aggravates neuronal degeneration and loss. a Accumulation of α-syn and neuroinflammation lead to degeneration and loss of DA neurons in substantia nigra. b The dead neurons release α-syn, which promotes activation of microglia, then the activated microglia release proinflammatory cytokines, which in turn aggravate neuronal death. c α-Syn promotes activation of microglia in several ways. Created with BioRender.com
Microglia degrade α-syn by autophagy and phagocytosis. Created with BioRender.com
Role of α-synuclein in microglia: autophagy and phagocytosis balance neuroinflammation in Parkinson’s disease

January 2023

·

303 Reads

·

45 Citations

Inflammation Research

Background Parkinson’s disease (PD) is the second most common neurodegenerative disease, and is characterized by accumulation of α-synuclein (α-syn). Neuroinflammation driven by microglia is an important pathological manifestation of PD. α-Syn is a crucial marker of PD, and its accumulation leads to microglia M1-like phenotype polarization, activation of NLRP3 inflammasomes, and impaired autophagy and phagocytosis in microglia. Autophagy of microglia is related to degradation of α-syn and NLRP3 inflammasome blockage to relieve neuroinflammation. Microglial autophagy and phagocytosis of released α-syn or fragments from apoptotic neurons maintain homeostasis in the brain. A variety of PD-related genes such as LRRK2, GBA and DJ-1 also contribute to this stability process. Objectives Further studies are needed to determine how α-syn works in microglia. Methods A keyword-based search was performed using the PubMed database for published articles. Conclusion In this review, we discuss the interaction between microglia and α-syn in PD pathogenesis and the possible mechanism of microglial autophagy and phagocytosis in α-syn clearance and inhibition of neuroinflammation. This may provide a novel insight into treatment of PD.


BAG3 promotes autophagy and suppresses NLRP3 inflammasome activation in Parkinson’s disease

November 2022

·

73 Reads

·

12 Citations

Annals of Translational Medicine

Background: Neuroinflammation mediated by microglia plays a key role in the pathogenesis of Parkinson's disease (PD), and our previous studies showed this was significantly inhibited by enhanced autophagy. In the autophagy pathway, Bcl2-associated athanogene (BAG)3 is a prominent co-chaperone, and we have shown BAG3 can regulate autophagy to clear the PD pathogenic protein α-synuclein. However, the connection between BAG3 and microglia mediated neuroinflammation is not clear. Methods: In this study, we explored whether BAG3 regulated related neuroinflammation and its original mechanism in PD. An inflammatory model of PD was established by injecting adeno-associated virus (AAV)-BAG3 into the bilateral striatum of C57BL/6 male mice to induce overexpression of BAG3, followed by injection of lipopolysaccharide (LPS). The striatum was extracted at 3 days after injection of LPS for Western blotting and reverse transcription quantitative polymerase chain reaction (RT-qPCR), and immunohistochemical staining was performed at 21 days after injection. At the same time, LPS was used to induce activation of BV2 cells to verify the effect of BAG3 in vitro. Results: Overexpression of BAG3 reduced LPS-induced pyroptosis by reducing activation of caspase-1, the NOD-like receptor family, and the pyrin domain-containing 3 (NLRP3) inflammasome, and by release of interleukin (IL)-1β and tumor necrosis factor (TNF)-α. The LPS-induced inflammatory environment inhibits autophagy, and overexpression of BAG3 can restore autophagy, which may be the mechanism by which BAG3 reduces neuronal inflammation in PD. Conclusions: Our results demonstrate BAG3 promotes autophagy and suppresses NLRP3 inflammasome formation in PD.

Citations (5)


... Studies using microglia and astrocytes isolated from TLR4-knockout mice showed that TLR4 is involved in α-Syn-induced activation of microglia and astrocytes but not in α-Syn internalization [31,76]. Melatonin receptor 1 regulated LC3-associated phagocytosis, which is involved in α-Syn clearance by microglia [77]. Further research is required to identify the cell surface receptors or binding sites in pericytes that interact with α-Syn. ...

Reference:

α-Synuclein Degradation in Brain Pericytes Is Mediated via Akt, ERK, and p38 MAPK Signaling Pathways
Microglial Melatonin Receptor 1 Degrades Pathological Alpha‐Synuclein Through Activating LC3‐Associated Phagocytosis In Vitro

... Studies using a mouse model with bilateral striatal injections of α-synuclein preformed fibrils (PFFs) have shown that α-synuclein aggregation induces iron deposition and ferroptosis, accompanied by increased lipid peroxidation and elevated free iron levels, particularly ferrous iron. When the melatonin receptor 1 (MT1) is knocked out, these changes are intensified, resulting in significant dopaminergic neuron loss and severe motor deficits [90]. Alzheimer's disease (AD) is primarily characterized by memory loss, cognitive decline, and behavioral changes. ...

Melatonin MT1 receptors regulate the Sirt1/Nrf2/Ho-1/Gpx4 pathway to prevent α-synuclein-induced ferroptosis in Parkinson's disease
  • Citing Article
  • March 2024

Journal of Pineal Research

... In PD, impaired HRV has been associated with disease severity, the duration of motor symptoms, and the dosage of dopaminergic medications [101,102]. Devos et al. [103] also observed progressive nocturnal cardiac dysregulation as PD advances. Specifically, the more advanced the PD, the lower the high-frequency (HF) HRV components and the higher the low-frequency/high-frequency (LF/HF) ratio. ...

Circadian disruption and sleep disorders in neurodegeneration

Translational Neurodegeneration

... Ceramide, a foundational sphingolipid responsible for the production of more intricate sphingolipids, is predominantly found in neurons where it regulates various neuronal functions such as signaling, synaptic transmission, metabolism, neuron-gliacommunication, and cell survival [39][40][41]. Dysfunctional autophagy has been linked to impaired removal of faulty mitochondria and α-synuclein, a key pathogenic mechanism in PD [42,43].Ceramide triggers the generation of activated lipids and autophagic markers within mitochondria, facilitating the targeting of mitochondria to autophagosomes and promoting ceramide-induced mitophagy [44,45]. The results of a corresponding animal experiment also supported our results. ...

Role of α-synuclein in microglia: autophagy and phagocytosis balance neuroinflammation in Parkinson’s disease

Inflammation Research

... Inhibition of SGK1 in glial cells can correct the inflammatory characteristics of glial cells by inhibiting the NF-κB-, NLRP3 inflammasome-, and cGAS-STING-mediated inflammatory pathways [96]. Similarly, activation of nuclear factor erythroid 2-related factor 2 (Nrf2) and JWA (a tumor suppressor gene), and overexpression of BAG3 (Bcl2-associated athanogene 3) can inhibit the activation of NLRP3 and caspase-1, reduce the nuclear transfer of NF-κB, and thereby suppress inflammation [97][98][99][100]. These findings collectively suggest that modulation of inflammasome activation in PD can be achieved by activating or inhibiting upstream molecules. ...

BAG3 promotes autophagy and suppresses NLRP3 inflammasome activation in Parkinson’s disease

Annals of Translational Medicine