Gaocai Li's research while affiliated with Huazhong University of Science and Technology and other places

What is this page?


This page lists the scientific contributions of an author, who either does not have a ResearchGate profile, or has not yet added these contributions to their profile.

It was automatically created by ResearchGate to create a record of this author's body of work. We create such pages to advance our goal of creating and maintaining the most comprehensive scientific repository possible. In doing so, we process publicly available (personal) data relating to the author as a member of the scientific community.

If you're a ResearchGate member, you can follow this page to keep up with this author's work.

If you are this author, and you don't want us to display this page anymore, please let us know.

Publications (52)


Schematic illustration of the Piezoelectric biomimetic bone-periosteum scaffold (TCP-PLA/GeSe) for spatiotemporal innervated bone regeneration and tumor ablation. Piezoelectric therapy of TCP-PLA/GeSe scaffold with ultrasound (US)-driven would effectively promote early neurogenic differentiation and osteogenesis. The synergistic effect of the piezoelectric effect and TCP bioceramic promotes long-term bone regeneration. Furthermore, the remarkable photothermal-conversion efficiency of the TCP-PLA/GeSe scaffold, coupled with its controlled Se release, enables exceptionally effective tumor eradication. This scaffold, TCP-PLA/GeSe, serves as a captivating biomaterial platform, offering multifunctional capabilities that hold great promise for diverse applications in spatiotemporal innervated bone regeneration and osteosarcoma treatment
Characterizations of GeSe nanosheets and TCP-PLA/GeSe scaffold. (A) Schematic illustration of the fabrication of TCP-PLA/GeSe scaffolds. (B) TEM images of GeSe nanosheets. (C) The Raman spectroscopy of GeSe nanosheets. (D) Temperature changing curves of GeSe nanosheets in vitro at different concentrations. (E) Temperature changing curves of GeSe nanosheets in vitro at different laser power densities of NIR. (F) Temperature profiles of scaffolds during four lasers on/off cycles. (G) Heating and cooling curve of the aqueous dispersions of GeSe nanosheets (1.0 mg/mL) under 808 nm (1.0 W/cm²) laser irradiation, and the linear fitting curve of time and -Ln (ɵ) acquired from the cooling period. (H-I) The SEM images of TCP-10%PLA/GeSe scaffold. (J) SEM image and element mapping of PLA/10%GeSe nanofiber membrane. (K) Temperature changing curves of TCP-PLA/GeSe scaffolds in vitro at different concentrations (L-M) The open-circuit voltage generated by TCP-10%PLA/GeSe scaffold under US excitation. (N-O) The short-circuit current generated by TCP-10%PLA/GeSe scaffold under US excitation
The neurogenic activity of SCs on TCP-PLA/GeSe scaffolds under US excitation. (A) The differentially expressed genes(GEGs) between TCP-PLA and TCP-PLA/GeSe + US group. (B) The heat map of gene expression for the neurogenic activity between the TCP-PLA and TCP-PLA/GeSe + US groups.(C) Top 20 KEGG pathways of genes enriched in the TCP-PLA/GeSe + US group. RT-PCR analysis of (D) BDNF and (E) NGF gene expression in different groups. (F) Representative images of BDNF proteins expression of SCs cultured in different groups. (G) Statistical analysis of BDNF expression of SCs cultured in different groups. (H) Representative images showing NGF proteins expression in SCs cultured under different conditions (I) Statistical analysis of NGF expression of SCs cultured in different groups. (J) Immunostaining images of PC12 cells cultured in SC-conditioned medium of different groups. (K) Statistical analysis of neurite length. (L) Schematic illustration of the mechanism of TCP-PLA/GeSe scaffold promoting neurogenic differentiation. n = 3. (* p < 0.05, ** p < 0.01, *** p < 0.001)
Osteogenic activity of BMSCs on TCP-PLA/GeSe scaffolds under US excitation. (A) Fluorescent images of BMSCs on TCP-PLA and TCP-PLA/GeSe scaffolds with/without US excitation. (B) Viability of BMSCs on TCP-PLA and TCP-PLA/GeSe scaffolds with/without US irritation. RT-PCR analysis of (C) OPN, (D) Runx2, (E) COL-1, and (F+) ALP gene expression in different groups. (G) Typical fluorescence pictures depicting OPN and Runx2 protein expression in different groups. (H) The statistical analysis of OPN and Runx2 expression in different groups. (I) ALP and ARS staining of BMSCs in different groups. (J) ALP and (K) ARS quantitative assay in different groups. n = 3. (* p < 0.05, ** p < 0.01, *** p < 0.001)
Performance of repairing large segmental bone defects of TCP-PLA/GeSe scaffolds under US excitation. (A) Large segmental bone defect in the middle radius of New Zealand rabbits. (B, C) Micro-CT quantification of newly formed bone. (D) 3D-reconstructed images of newly formed bone in different groups. (E) The regeneration of bone was evaluated 12 weeks after surgery using H&E and (F) Masson stains. n = 3. (* p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001)

+2

Biomimetic bone-periosteum scaffold for spatiotemporal regulated innervated bone regeneration and therapy of osteosarcoma
  • Article
  • Full-text available

May 2024

·

33 Reads

Journal of Nanobiotechnology

Yan Xu

·

Chao Xu

·

Huan Song

·

[...]

·

The complexity of repairing large segment defects and eradicating residual tumor cell puts the osteosarcoma clinical management challenging. Current biomaterial design often overlooks the crucial role of precisely regulating innervation in bone regeneration. Here, we develop a Germanium Selenium (GeSe) co-doped polylactic acid (PLA) nanofiber membrane-coated tricalcium phosphate bioceramic scaffold (TCP-PLA/GeSe) that mimics the bone-periosteum structure. This biomimetic scaffold offers a dual functionality, combining piezoelectric and photothermal conversion capabilities while remaining biodegradable. When subjected to ultrasound irradiation, the US-electric stimulation of TCP-PLA/GeSe enables spatiotemporal control of neurogenic differentiation. This feature supports early innervation during bone formation, promoting early neurogenic differentiation of Schwann cells (SCs) by increasing intracellular Ca²⁺ and subsequently activating the PI3K-Akt and Ras signaling pathways. The biomimetic scaffold also demonstrates exceptional osteogenic differentiation potential under ultrasound irradiation. In rabbit model of large segment bone defects, the TCP-PLA/GeSe demonstrates promoted osteogenesis and nerve fibre ingrowth. The combined attributes of high photothermal conversion capacity and the sustained release of anti-tumor selenium from the TCP-PLA/GeSe enable the synergistic eradication of osteosarcoma both in vitro and in vivo. This strategy provides new insights on designing advanced biomaterials of repairing large segment bone defect and osteosarcoma. Supplementary Information The online version contains supplementary material available at 10.1186/s12951-024-02430-7.

Download
Share

Altered Metabolism and Inflammation Driven by Post-translational Modifications in Intervertebral Disc Degeneration

April 2024

·

32 Reads

Research

Research

Intervertebral disc degeneration (IVDD) is a prevalent cause of low back pain and a leading contributor to disability. IVDD progression involves pathological shifts marked by low-grade inflammation, extracellular matrix remodeling, and metabolic disruptions characterized by heightened glycolytic pathways, mitochondrial dysfunction, and cellular senescence. Extensive posttranslational modifications of proteins within nucleus pulposus cells and chondrocytes play crucial roles in reshaping the intervertebral disc phenotype and orchestrating metabolism and inflammation in diverse contexts. This review focuses on the pivotal roles of phosphorylation, ubiquitination, acetylation, glycosylation, methylation, and lactylation in IVDD pathogenesis. It integrates the latest insights into various posttranslational modification-mediated metabolic and inflammatory signaling networks, laying the groundwork for targeted proteomics and metabolomics for IVDD treatment. The discussion also highlights unexplored territories, emphasizing the need for future research, particularly in understanding the role of lactylation in intervertebral disc health, an area currently shrouded in mystery.


Disassembly of the TRIM56-ATR complex promotes cytoDNA/cGAS/STING axis–dependent intervertebral disc inflammatory degeneration

January 2024

·

27 Reads

·

1 Citation

The Journal of clinical investigation

As the leading cause of disability worldwide, low back pain (LBP) is recognized as a pivotal socioeconomic challenge to the aging population and is largely attributed to intervertebral disc degeneration (IVDD). Elastic nucleus pulposus (NP) tissue is essential for the maintenance of IVD structural and functional integrity. The accumulation of senescent NP cells with an inflammatory hypersecretory phenotype due to aging and other damaging factors is a distinctive hallmark of IVDD initiation and progression. In this study, we reveal a mechanism of IVDD progression in which aberrant genomic DNA damage promoted NP cell inflammatory senescence via activation of the cyclic GMP-AMP synthase/stimulator of IFN genes (cGAS/STING) axis but not of absent in melanoma 2 (AIM2) inflammasome assembly. Ataxia-telangiectasia-mutated and Rad3-related protein (ATR) deficiency destroyed genomic integrity and led to cytosolic mislocalization of genomic DNA, which acted as a powerful driver of cGAS/STING axis-dependent inflammatory phenotype acquisition during NP cell senescence. Mechanistically, disassembly of the ATR-tripartite motif-containing 56 (ATR-TRIM56) complex with the enzymatic liberation of ubiquitin-specific peptidase 5 (USP5) and TRIM25 drove changes in ATR ubiquitination, with ATR switching from K63- to K48-linked modification, c thereby promoting ubiquitin-proteasome-dependent dynamic instability of ATR protein during NP cell senescence progression. Importantly, an engineered extracellular vesicle-based strategy for delivering ATR-overexpressing plasmid cargo efficiently diminished DNA damage-associated NP cell senescence and substantially mitigated IVDD progression, indicating promising targets and effective approaches to ameliorate the chronic pain and disabling effects of IVDD.


The NLRX1-SLC39A7 complex orchestrates mitochondrial dynamics and mitophagy to rejuvenate intervertebral disc by modulating mitochondrial Zn2+ trafficking

October 2023

·

18 Reads

·

6 Citations

Intervertebral disc degeneration (IDD) is the most critical pathological factor in the development of low back pain. The maintenance of nucleus pulposus (NP) cell and intervertebral disc integrity benefits largely from well-controlled mitochondrial quality, surveilled by mitochondrial dynamics (fission and fusion) and mitophagy, but the outcome is cellular context-dependent that remain to be clarified. Our studies revealed that the loss of NLRX1 is correlated with NP cell senescence and IDD progression, which involve disordered mitochondrial quality. Further using animal and in vitro tissue and cell models, we demonstrated that NLRX1 could facilitate mitochondrial quality by coupling mitochondrial dynamic factors (p-DNM1L, L-OPA1:S-OPA1, OMA1) and mitophagy activity. Conversely, mitochondrial collapse occurred in NLRX1-defective NP cells and switched on the compensatory PINK1-PRKN pathway that led to excessive mitophagy and aggressive NP cell senescence. Mechanistically, NLRX1 was originally shown to interact with zinc transporter SLC39A7 and modulate mitochondrial Zn2+ trafficking via the formation of an NLRX1-SLC39A7 complex on the mitochondrial membrane of NP cells, subsequently orchestrating mitochondrial dynamics and mitophagy. The restoration of NLRX1 function by gene overexpression or pharmacological agonist (NX-13) treatment showed great potential for regulating mitochondrial fission with synchronous fusion and mitophagy, thus sustaining mitochondrial homeostasis, ameliorating NP cell senescence and rejuvenating intervertebral discs. Collectively, our findings highlight a working model whereby the NLRX1-SLC39A7 complex coupled mitochondrial dynamics and mitophagy activity to surveil and target damaged mitochondria for degradation, which determines the beneficial function of the mitochondrial surveillance system and ultimately rejuvenates intervertebral discs.Abbreviations: 3-MA: 3-methyladenine; Baf-A1: bafilomycin A1; CDKN1A/p21: cyclin dependent kinase inhibitor 1A; CDKN2A/p16: cyclin dependent kinase inhibitor 2A; DNM1L/DRP1: dynamin 1 like; EdU: 5-Ethynyl-2'-deoxyuridine; HE: hematoxylin-eosin; IDD: intervertebral disc degeneration; IL1B/IL-1β: interleukin 1 beta; IL6: interleukin 6; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MKI67/Ki67: marker of proliferation Ki-67; LBP: low back pain; MMP: mitochondrial membrane potential; MFN1: mitofusin 1; MFN2: mitofusin 2; MFF: mitochondrial fission factor; NP: nucleus pulposus; NLRX1: NLR family member X1; OMA1: OMA1 zinc metallopeptidase; OPA1: OPA1 mitochondrial dynamin like GTPase; PINK1: PTEN induced kinase 1; PRKN: parkin RBR E3 ubiquitin protein ligase; ROS: reactive oxidative species; SASP: senescence-associated secretory phenotype; SA-GLB1/β-gal: senescence-associated galactosidase beta 1; SO: safranin o; TBHP: tert-butyl hydroperoxide; TP53/p53: tumor protein p53; SLC39A7/ZIP7: solute carrier family 39 member 7; TOMM20: translocase of outer mitochondrial membrane 20; TIMM23: translocase of inner mitochondrial membrane 23.


Matrix stiffness induces Drp1-mediated mitochondrial fission through Piezo1 mechanotransduction in human intervertebral disc degeneration

October 2023

·

64 Reads

·

2 Citations

Journal of Translational Medicine

Background Extracellular matrix stiffness is emerging as a crucial mechanical cue that drives the progression of various diseases, such as cancer, fibrosis, and inflammation. The matrix stiffness of the nucleus pulposus (NP) tissues increase gradually during intervertebral disc degeneration (IDD), while the mechanism through which NP cells sense and react to matrix stiffness remains unclear. In addition, mitochondrial dynamics play a key role in various cellular functions. An in-depth investigation of the pathogenesis of IDD can provide new insights for the development of effective therapies. In this study, we aim to investigate the effects of matrix stiffness on mitochondrial dynamics in IDD. Methods To build the gradient stiffness model, NP cells were cultured on polystyrene plates with different stiffness. Western blot analysis, and immunofluorescence staining were used to detect the expression of mitochondrial dynamics-related proteins. Flow cytometry was used to detect the mitochondrial membrane potential and intracellular Ca²⁺ levels. Apoptosis related proteins, ROS level, and TUNEL staining were performed to assess the effect of substrate stiffness on NP cells. Results Stiff substrate increased phosphorylation of dynamin-related protein 1 (Drp1) at Ser616 by activating extracellular signal-regulated kinase 1/2 (ERK1/2) pathway, which promoted mitochondrial fission and apoptosis in NP cells. Furthermore, Piezo1 activation was involved in the regulation of the post-translational modifications of Drp1 and mitochondrial fission caused by matrix stiffness. Inhibition of Piezo1 and ERK1/2 can effectively reduce stiffness-induced ROS elevation and apoptosis in NP cells. Conclusions Our results revealed that stiff substrate causes Piezo1 activation and Ca²⁺ influx, results in ERK1/2 activation and phosphorylation of Drp1 at S616, and finally leads to mitochondrial fission and apoptosis in NP cells. These findings reveal a new mechanism of mechanotransduction in NP cells, providing novel insights into the development of therapies for treating IDD.


Fig. 2 Impaired TFEB-regulated lysosomal dysregulation contributes to NPC senescence. a Representative images and quantification of total LysoTracker Red in NPCs with the indicated treatment (Scale bar: 10 μm). b Representative images and quantification of Magic Red dye in NPCs with the indicated treatment (Scale bar: 10 μm). c IF staining of TFEB in NPCs with the indicated treatment and the quantification of TFEB expression in the nucleus labeled with DAPI (Scale bar: 10 μm). d Cellular fractionation analysis of TFEB in NPCs with the indicated treatment. e Western blot analysis of p-TFEB and TFEB in human NPCs with the indicated treatment. f Western blot analysis of p53, p21, p16 in human NPCs transfected with si-TFEB. g, h Western blot analysis of LC3/p62 in human NPCs transfected with si-TFEB. i IF staining of TFEB in NPCs with the indicated treatment (Scale bar: 10 μm). j RT-qPCR analysis for TFEB target genes (TPP1, CTSF, MCOLN1, CTSD, LAMP1, ATG9A, WIPI1, ATG10, LC3B, SQSTM1) in NPCs with the indicated treatment. k Western blot analysis of p53, p21, p16 in human NPCs with the indicated treatment. l SA-β-gal activity staining and analysis of NPCs with the indicated treatment (Scale bar: 100 μm). m EdU incorporation assay and analysis of NPCs with the indicated treatment (Scale bar: 100 μm). Data are expressed as mean ± SD. * p < 0.05, ** p < 0.01, ** p < 0.001, **** p < 0.0001, ns not significant, two-tailed unpaired t-test and and one-way ANOVA.
Fig. 4 K141 methylation of PPP1CA disrupts its binding with TFEB. a Schematic workflow showing TFEB Co-IP with NPCs followed by LC-MS/MS experiments. b LC-MS/MS spectrum of the tryptic peptide IYGFYDECKR, carrying a mass of +14.0156 Da at residues Lys141. c, d Exogenous Co-IP analysis of the interaction of TFEB with PP1 in HEK293T cells cotransfected with Flag-TFEB and His-PPP1CA. e, f Endogenous Co-IP analysis of the interaction of TFEB with PPP1CA in NPCs. g Cellular fractionation analysis of TFEB in NPCs transfected with si-PPP1CA. h Western blot analysis of p-TFEB and TFEB in human NPCs transfected with si-PPP1CA. (i) IF staining of TFEB in NPCs transfected with si-PPP1CA and the quantification of TFEB expression in the nucleus labeled with DAPI (Scale bar: 10 μm). j Sequence alignment of PPP1CA across multiple species near the K141. k Co-IP analysis of methylated PPP1CA in NPCs with the indicated treatment. l Co-IP analysis of methylated PPP1CA in NPCs transfected with PPP1CA His-tagged wild-type PPP1CA, KR mutants and KM mutants. m IF analysis of the interaction of TFEB with PPP1CA in NPCs treated with the indicated treatment (Scale bar: 10 μm, 2μm). n Co-IP analysis of the interaction of TFEB with PPP1CA in NPCs with the indicated treatment. o Co-IP analysis of the interaction of TFEB with PPP1CA in NPCs cotransfected with Flag-TFEB and His-PPP1CA (wild-type, KR mutants) with the indicated treatment. p Co-IP analysis of the interaction of TFEB with PPP1CA in HEK293T cells cotransfected with Flag-TFEB and His-PPP1CA (wild-type, KR mutants and KM mutants). Data are expressed as mean ± SD. * p < 0.05, ns not significant, two-tailed unpaired t test.
Lysine methylation of PPP1CA by the methyltransferase SUV39H2 disrupts TFEB-dependent autophagy and promotes intervertebral disc degeneration

August 2023

·

50 Reads

·

3 Citations

Cell Death and Differentiation

Impaired transcription factor EB (TFEB) function and deficient autophagy activity have been shown to aggravate intervertebral disc (IVD) degeneration (IDD), yet the underlying mechanisms remain less clear. Protein posttranslational modifications (PTMs) are critical for determining TFEB trafficking and transcriptional activity. Here, we demonstrate that TFEB activity is controlled by protein methylation in degenerated nucleus pulposus cells (NPCs), even though TFEB itself is incapable of undergoing methylation. Specifically, protein phosphatase 1 catalytic subunit alpha (PPP1CA), newly identified to dephosphorylate TFEB, contains a K141 mono-methylated site. In degenerated NPCs, increased K141-methylation of PPP1CA disrupts its interaction with TEFB and subsequently blocks TEFB dephosphorylation and nuclear translocation, which eventually leads to autophagy deficiency and NPC senescence. In addition, we found that the PPP1CA-mediated targeting of TFEB is facilitated by the protein phosphatase 1 regulatory subunit 9B (PPP1R9B), which binds with PPP1CA and is also manipulated by K141 methylation. Further proteomic analysis revealed that the protein lysine methyltransferase suppressor of variegation 3-9 homologue 2 (SUV39H2) is responsible for the K141 mono-methylation of PPP1CA. Targeting SUV39H2 effectively mitigates NPC senescence and IDD progression, providing a potential therapeutic strategy for IDD intervention.


Back Cover: Recent advances of cellular stimulation with triboelectric nanogenerators (EXP2 4/2023)

August 2023

·

37 Reads


Sonodynamic amplification of cGAS-STING activation by cobalt-based nanoagonist against bone and metastatic tumor

August 2023

·

31 Reads

·

4 Citations

Biomaterials

The therapeutic effect of cancer immunotherapy is restrained by limited patient response rate caused by 'cold' tumors with an intrinsically immunosuppressive tumor microenvironment (TME). Activating stimulator of interferon genes (STING) confers promising antitumor immunity even in 'cold' tumors, but the further promotion of STING agonists is hindered by undesirable toxicity, low specificity and lack of controllability. Herein, an ultrasound-controllable cGAS-STING amplifying nanoagonist was constructed by coordinating mitochondria-targeting ligand triphenylphosphonium (TPP) to sonodynamic cobalt organic framework nanosheets (TPP@CoTCPP). The Co ions specifically amplify STING activation only when cytosolic mitochondrial DNA leakage is caused by sonocatalysis-induced ROS production and sensed by cGAS. A series of downstream innate immune proinflammatory responses induced by local cGAS-STING pathway activation under spatiotemporal ultrasound stimulation efficiently prime the antitumor T-cell response against bone metastatic tumor, a typical immunosuppressive tumor. We also found that the coordination of TPP augments the sonodynamic effect of CoTCPP nanosheets by reducing the band gap, improving O2 adsorption and enhancing electron transfer. Overall, our study demonstrates that the targeted and amplified cGAS-STING activation in cancer cell controlled by spatiotemporal ultrasound irradiation boosts high-efficiency sonodynamic-ionicimmunotherapy against immunosuppressive tumor.


Copper Ion‐Modified Germanium Phosphorus Nanosheets Integrated with an Electroactive and Biodegradable Hydrogel for Neuro‐Vascularized Bone Regeneration

July 2023

·

45 Reads

·

11 Citations

Severe bone defects accompanied by vascular and peripheral nerve injuries represent a huge orthopedic challenge and are often accompanied by the risk of infection. Thus, biomaterials with antibacterial and neurovascular regeneration properties are highly desirable. Here, a newly designed biohybrid biodegradable hydrogel (GelMA) containing copper ion‐modified germanium‐phosphorus (GeP) nanosheets, which act as neuro‐vascular regeneration and antibacterial agents, is designed. The copper ion modification process serves to improve the stability of the GeP nanosheets and offers a platform for the sustained release of bioactive ions. Study findings show that GelMA/GeP@Cu has effective antibacterial properties. The integrated hydrogel can significantly boost the osteogenic differentiation of bone marrow mesenchymal stem cells, facilitate angiogenesis in human umbilical vein endothelial cells, and up‐regulate neural differentiation‐related proteins in neural stem cells in vitro. In vivo, in the rat calvarial bone defect mode, the GelMA/GeP@Cu hydrogel is found to enhance angiogenesis and neurogenesis, eventually contributing to bone regeneration. These findings indicate that in the field of bone tissue engineering, GelMA/GeP@Cu can serve as a valuable biomaterial for neuro‐vascularized bone regeneration and infection prevention.


Recent advances of cellular stimulation with triboelectric nanogenerators

May 2023

·

57 Reads

·

8 Citations

Exploration

Exploration

Abstract Triboelectric nanogenerators (TENGs) are new energy collection devices that have the characteristics of high efficiency, low cost, miniaturization capability, and convenient manufacture. TENGs mainly utilize the triboelectric effect to obtain mechanical energy from organisms or the environment, and this mechanical energy is then converted into and output as electrical energy. Bioelectricity is a phenomenon that widely exists in various cellular processes, including cell proliferation, senescence, apoptosis, as well as adjacent cells’ communication and coordination. Therefore, based on these features, TENGs can be applied in organisms to collect energy and output electrical stimulation to act on cells, changing their activities and thereby playing a role in regulating cellular function and interfering with cellular fate, which can further develop into new methods of health care and disease intervention. In this review, we first introduce the working principle of TENGs and their working modes, and then summarize the current research status of cellular function regulation and fate determination stimulated by TENGs, and also analyze their application prospects for changing various processes of cell activity. Finally, we discuss the opportunities and challenges of TENGs in the fields of life science and biomedical engineering, and propose a variety of possibilities for their potential development direction.


Citations (44)


... Additionally, in the literature, using live mammalian subjects and in vitro tissue and cell models, it is mentioned that nucleotidebinding oligomerization domain (NOD)-like receptor family member X1 (NLRX1) can facilitate mitochondrial quality by combining mitophagy activity and mitochondrial dynamic factors such as dynamin 1-like protein, which is involved in the division of mitochondria and peroxisomes. It is reported that in NP cells with NLRX1 damage, mitochondrial collapse occurs and the compensatory PINK1-Parkin RBR E3 Ubiquitin Protein Ligaz (PRKN) signaling pathway is activated, leading to excessive mitophagy and aggressive NP cell senescence (28) . NLRX1 has been shown to initially interact with the zinc transporter SLC39A7 and modulate mitochondrial Zn2+ through the formation of an NLRX1-SLC39A7 complex on the mitochondrial membrane of NP cells, subsequently regulating mitochondrial dynamics and mitophagy and improving IVD (28) . ...

Reference:

COULD PHARMACOLOGICAL TARGETING MITOPHAGIC OR LYSOPHAGIC SIGNALING PATHWAYS BE A NEW HOPE IN THE TREATMENT OF INTERVERTEBRAL DISC DEGENERATION?
The NLRX1-SLC39A7 complex orchestrates mitochondrial dynamics and mitophagy to rejuvenate intervertebral disc by modulating mitochondrial Zn2+ trafficking
Autophagy

Autophagy

... Importantly, centering the evaluation on mechanotransduction it is possible to appreciate the contribution of physical cues in tuning pharmacological activity which is typically not included in in vitro testing, but can represent a major confounder for bench-to-bedside translation of experimental results. [46,47] was purchased from abcam (Cambridge, UK). Solid substances were dissolved in DMSO or water as described by the supplier. ...

Matrix stiffness induces Drp1-mediated mitochondrial fission through Piezo1 mechanotransduction in human intervertebral disc degeneration

Journal of Translational Medicine

... Therefore, the prepared functional 3DTi by Cu ion implantation combined with UV photofunctionalization can effectively promote bone formation. [27]. Therefore, the induction of angiogenesis of the proposed implants was examined. ...

Copper Ion‐Modified Germanium Phosphorus Nanosheets Integrated with an Electroactive and Biodegradable Hydrogel for Neuro‐Vascularized Bone Regeneration
Advanced Healthcare Materials

Advanced Healthcare Materials

... At present, the pathological mechanism leading to IDD remains to be studied, and ferroptosis is one of the pathological mechanisms leading to IDD [36,37]. Ferroptosis is a newly discovered form of cell death that is characterized by the accumulation of lipid peroxidation and irondependent reactive oxygen species (ROS) [38]. ...

Stiff Substrate Induces Nucleus Pulposus Cell Ferroptosis via YAP and N‐Cadherin Mediated Mechanotransduction
Advanced Healthcare Materials

Advanced Healthcare Materials

... Tumor-derived EVs mediate bidirectional communication between tumor and stromal cells in local and distant microenvironments. Tumor-derived EVs activate stromal fibroblasts to differentiate into cancer-associated fibroblasts (CAFs), which supports tumor growth, stimulates endothelial cells to increase angiogenesis and induces the suppression of the antitumor immune response, thereby generating a favorable premetastatic microenvironment (14). Adaptation to the distant microenvironment is critical for metastasizing at different sites or organs. ...

Interactions between extracellular vesicles and microbiome in human diseases: New therapeutic opportunities

... The modified EVs restored the cellular uptake rate, delivered Prx2 into NP cells and inhibited TNF-αinduced pyroptosis. In another study, Tong et al. [34] investigated whether the delivery efficiency of MSC-EVs was decreased in NP cells under hypoxic conditions because of an increase in endocytic recycling. Hypoxia inducible factor-1 (HIF-1)-induced upregulation of Rabcoupling protein (RCP), which is also known as RAB11 family interacting protein 1 (RAB11FIP1), promoted the Rab11a-dependent recycling of internalized MSC-EVs by facilitating the interaction between Rab11a and MSC-EVs. ...

Augmenting Intracellular Cargo Delivery of Extracellular Vesicles in Hypoxic Tissues through Inhibiting Hypoxia-Induced Endocytic Recycling
  • Citing Article
  • February 2023

ACS Nano

... Epigenetic adjustments can be induced by simple cell deformations, for example in response to the surrounding matrix in animals (Alisafaei et al., 2019) or to prolonged mechanical stimulation in plants (Brenya et al., 2022). Together, these gave rise to the model of the mechanical priming of cell identity and stress resistance via epigenetic processes (Brenya et al., 2022;Li et al., 2022). ...

Mechanoceutical forces squeeze the epigenetic changes
  • Citing Article
  • October 2022

Matter

... Due to its unique advantages, this technology has gained significant attention in the field. Similarly, various bioactive compounds possess the capability to effectively penetrate the NP by delivering biomaterials, thereby facilitating the replenishment of damaged NP tissues, alleviating the inflammatory microenvironment within NP, rectifying disrupted ECM metabolism, and ultimately restoring the structural and mechanical properties of intervertebral discs (Wu et al., 2022). Song et al. have published a concise review article on bioactive substances that possess the potential to delay and reverse the progression of IVDD. ...

Repair Strategies and Bioactive Functional Materials for Intervertebral Disc

... Unfortunately, the study lacked normal intervertebral disc tissue samples as a control. However, Luo et al. (2022) found that OGT expression and O-GlcNAcylation were elevated in degenerated nucleus pulposus tissue and nutritionally deficient nucleus pulposus cells compared to nucleus pulposus cells in healthy disc tissue. Furthermore, inhibition of O-GlcNAcylation promoted the induction of apoptosis and senescence in human nucleus pulposus cells by nutrient deficiency. ...

O-GlcNAc transferase regulates intervertebral disc degeneration by targeting FAM134B-mediated ER-phagy

Experimental and Molecular Medicine

... 7 Disk degeneration occurs consistently with advancing age as a result of disordered metabolism of the extracellular matrix (ECM) and a loss of healthy disk cells. 6,8,9 More specifically, abnormal catabolism driven by increased expression of inflammatory cytokines, including interleukin-1 beta (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor alpha (TNF-α), as well as catabolic mediators such as the matrix metalloproteinases (MMPs) family, is the main characteristic of IVDD. [10][11][12] Autophagy is believed to be a cytoprotective process through which intracellular proteins and organelles can be renewed and mobilized to fulfill the bioenergetic needs of the cell by generating energyrich compounds. ...

Epigenetic regulation in intervertebral disc degeneration
  • Citing Article
  • August 2022

Trends in Molecular Medicine