Codelivery of minocycline hydrochloride and dextran sulfate via bionic liposomes for the treatment of spinal cord injury

https://doi.org/10.1016/j.ijpharm.2022.122285Get rights and content

Abstract

After primary injury to the spinal cord, a series of microenvironmental changes can lead to secondary injury. The use of nano-targeted drug delivery systems to improve the postinjury microenvironment, inhibit inflammation and reduce neuronal apoptosis can be of great help during spinal cord injury (SCI) recovery. In this study, we prepared primary macrophage membranes bionic modified nanoliposomes (MH-DS@M−Lips) loaded with minocycline hydrochloride (MH) and dextran sulfate (DS) to target their delivery to the site of injury to bind calcium ions in situ and form metal ion complexes. Complex formation reduced calcium ion concentrations and calcium-associated neuronal apoptosis, while MH was slowly released to produce better anti-inflammatory effects. The successful preparation of MH-DS@M−Lips was verified using transmission electron microscopy (TEM), confocal laser scanning microscopy (CLSM), western blotting and dynamic light scattering (DLS). The targeting capability of the MH-DS@M−Lips was demonstrated using a Transwell system and an in vivo imaging system. The therapeutic efficacy of MH-DS@M−Lips was examined in vitro and in vivo using flow cytometry, immunofluorescence, ELISA kits and western blotting. The results showed that SCI mice treated with MH-DS@M−Lips received high behavioral scores, which led to the conclusion that MH-DS@M−Lips have great potential for the treatment of SCI.

Introduction

SCI is a serious traumatic disorder of the central nervous system (CNS). More than one million new cases of SCI occur each year worldwide as a result of accidents (Charlifue et al., 2016, Li et al., 2019). These injuries result in motor and sensory dysfunction (da Silva et al., 2020), and current clinical treatment strategies are not very effective during recovery (Hachem et al., 2017, Toluse and Adeyemi, 2021), leading to patients incurring lifelong treatment costs (Nogueira et al., 2012).

The main clinical drug treatment for SCI is high-dose systemic administration of methylprednisolone, which has been reported to have the therapeutic effects of reducing inflammation and neuroprotection (Cheung et al., 2015). The blood-spinal cord barrier (BSCB) protects CNS homeostasis and prevents the entry of foreign substances (Jin et al., 2021). Although the BSCB opens briefly at the injury site after SCI, the BSCB still poses a great challenge for subsequent drug delivery (Whetstone et al., 2003). Because of the BSCB, the systemic administration of conservative doses of methylprednisolone is inefficient, difficult to enrich at the injury site, and therapeutically ineffective (Jin et al., 2021, Kim et al., 2009), while the toxicity that may be triggered by high doses can cause serious and unavoidable damage to the body, such as gastric bleeding, liver damage and reinfection (Ersayli et al., 2006). In practice, the BSCB makes it difficult to systemically administer most drugs at safe doses, so it is worth exploring nanodelivery systems for delivering drugs across the BSCB to target the damaged areas (Song et al., 2019). In recent years, nanodelivery systems such as liposomes (Gao et al., 2017), PLGA nanoparticles (Ren et al., 2014), silica nanoparticles (Zhang et al., 2021), metal nanoparticles (Kim et al., 2017), exosomes (Guo et al., 2019) and cell membrane vesicles (Liu et al., 2022) have been reported.

Among these, nanoliposomes as a drug delivery system are less toxic, simple to prepare and have a proven process for mass production (Li et al., 2018, Wang et al., 2021). Our previous studies have shown that macrophage-derived membrane nanovesicles have a good effect to target the site of SCI (Liu et al., 2022). Therefore, we performed primary macrophage membranes bionic modified nanoliposomes. The modification method was different from that of the previously reported membrane-modified bionanoparticles (Tang et al., 2021b, Zhuang et al., 2020), as it used a one-step method to dope a small amount of the primary macrophage membranes into lipids, which could reduce the number of membranes needed and solve the problem of the limited source of bionanomimetic materials. Additionally, as less bionanomimetic materials were used, the possible unknown side effects of these materials were reduced. It was found that the modified bionic nanoliposomes inherited primary macrophage membranes proteins, such as VLA4 (Integrins α4/β1) and Mac-1 (CD11b/CD18). These proteins could interact with the highly expressed vascular cell adhesion molecule 1 (VCAM-1) or intercellular adhesion molecules (ICAM) in endothelial cells of the inflammatory site (David and Kroner, 2011, Thawer et al., 2013), which could make the bionic nanoliposomes target and enrich in the damaged area.

Following primary injury to the spinal cord caused by external forces, the microenvironment around the injury point rapidly undergoes a series of changes leading to secondary injury, including M1 macrophage infiltration causing hyperinflammation, ROS stress, electrolyte disturbances and elevated intra- and extracellular calcium ion concentrations (Ahuja et al., 2017). Inflammation and elevated levels of calcium ions lead to an increase in peripheral neuron apoptosis (Beattie, 2004). Improving the microenvironment at this point (e.g., reducing calcium ion concentrations) can help tremendously in SCI recovery (Schanne et al., 1979, Wang et al., 2017). Inflammatory regulation and neuroprotection have a significant impact on subsequent motor recovery, which should be prominently considered (Ahuja et al., 2017, Beattie, 2004). MH, a broad-spectrum antibacterial tetracycline antibiotic, has been reported to be able to treat SCI and possesses a powerful anti-inflammatory effect (Kobayashi et al., 2013). According to previous reports, MH and DS can form complexes with divalent metal ions such as calcium ions and magnesium ions (Wang et al., 2017, Zhang et al., 2015). We attempted to load both MH and DS in macrophage membranes modified nanobionic liposomes (MH-DS@M−Lips). MH-DS@M−Lips were constructed to hopefully target the site of injury for drug enrichment, reduce calcium ion concentrations in situ, reduce neuronal apoptosis and produce anti-inflammatory effects. Experiments simulating the in vivo damage environment showed that MH and DS bind to calcium ions, producing metal ion complexes (Fig. S1) similar to those previously reported (Zhang et al., 2015). This system was studied both in vitro and in vivo, and it was found that MH combined with DS reduced the subsequent calcium-associated apoptotic cascade by binding large amounts of free calcium ions, resulting in neuroprotection while MH produced an anti-inflammatory effect. SCI mice treated with MH-DS@M−Lips also achieved higher behavioral scores. The design concept and mechanism of action are shown in Scheme 1, which will hopefully provide new ideas for the treatment of SCI.

Section snippets

Materials

98 % minocycline hydrochloride (MH), M.W 5,000 dextran sulphate (DS), 99 % cholesterol were purchased from Aladdin Chemical Reagents. 98 % soy phosphatidylcholine were purchased from Shanghai Acmec Biochemical. Povidone iodine, starch broth, dulbecco's modification of eagle's medium dulbecco (DMEM), fetal bovine serum (FBS), penicillin–streptomycin, trypsin, phenylmethanesulfonyl fluoride (PMSF), lipopolysaccharide (LPS), 30 % H2O2, phosphate buffered saline (PBS), normal saline (NS),

Characterization of MH-DS@M−Lips

Immunofluorescence and flow cytometry were used to confirm that the extracted primary cells were macrophages. The cells were marked with the macrophage marker anti-F4/80 as determined by immunofluorescence (Fig. 1A), and flow cytometry showed that 99.31 % of the cells expressed F4/80 (Fig. 1B). The extracted cell membranes were observed using TEM (Fig. S2), and showed an irregular morphology. And TEM was also used to observe the microstructure of the MH-DS@M−Lips, the results showed that they

Discussion

In SCI, secondary injury is a very large obstacle to recovery that is caused by a series of postinjury microenvironmental changes, such as a calcium ion surge, oxidative stress, an increase in proinflammatory factors and the activation of apoptotic factors (Ahuja et al., 2017, Quadri et al., 2020). Therefore, in terms of SCI treatment strategies, postinjury microenvironmental modulation can reduce secondary damage, which is beneficial for SCI recovery (Wang et al., 2017). However, the presence

Conclusion

In this study, primary macrophage membranes bionic nanoliposomes loaded with MH and DS (MH-DS@M−Lips) were prepared for the treatment of SCI. MH-DS@M−Lips are simple to prepare, have good biosafety and stability, and can target inflammation. The in vitro cellular and in vivo animal experiments demonstrated the anti-inflammatory, anti-calcium ion surge, anti-ROS and neuroprotective effects of MH-DS@M−Lips. After treatment with MH-DS@M−Lips, there was a significant recovery of motor function in

Data availability statement

The raw data required to reproduce these findings cannot be shared at this time as the data also forms part of an ongoing study. The processed data (used in this manuscript) required to reproduce these findings can be shared at this time through personal request.

CRediT authorship contribution statement

Jinyu An: Conceptualization, Investigation, Methodology, Software, Validation, Writing – original draft, Formal analysis. Xue Jiang: Visualization, Methodology. Zhe Wang: Software, Formal analysis. Yingqiao Li: Software, Methodology. Zhiru Zou: Methodology. Qian Wu: Methodology. Le Tong: Methodology. Xifan Mei: Resources, Supervision, Funding acquisition. He Tian: Supervision. Chao Wu: Conceptualization, Resources, Supervision, Funding acquisition, Writing – review & editing.

Declaration of Competing Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgements

The authors acknowledge the financial support received from the Natural Science Foundation of Liaoning Province [No. 20180550155, 2021-MS-332], the National Natural Science Foundation of China (No. 81871556, 82072165), LiaoNing Revitalization Talents Program (No. XLYC1902108), Liaoning Provincial Key Laboratory of Marine Bioactive Substances and Technological Innovation Center of Liaoning Pharmaceutical Action and Quality Evaluation (No. 2020-10, No.2021-5).

References (57)

  • Y.H. Song et al.

    Recent advances in nanotherapeutic strategies for spinal cord injury repair

    Adv. Drug Deliv. Rev.

    (2019)
  • W. Tang et al.

    Macrophage membrane-mediated targeted drug delivery for treatment of spinal cord injury regardless of the macrophage polarization states

    Asian J. Pharm. Sci.

    (2021)
  • S.G. Thawer et al.

    Temporal changes in monocyte and macrophage subsets and microglial macrophages following spinal cord injury in the Lys-Egfp-ki mouse model

    J. Neuroimmunol.

    (2013)
  • Z. Wang et al.

    Local delivery of minocycline from metal ion-assisted self-assembled complexes promotes neuroprotection and functional recovery after spinal cord injury

    Biomaterials

    (2017)
  • X. Wang et al.

    Engineered liposomes targeting the gut-CNS Axis for comprehensive therapy of spinal cord injury

    J. Controlled Release: Official J. Controlled Release Soc.

    (2021)
  • B. Zhang et al.

    Macrophage-mediated degradable gelatin-coated mesoporous silica nanoparticles carrying pirfenidone for the treatment of rat spinal cord injury

    Nanomed. Nanotechnol. Biol. Med.

    (2021)
  • C.S. Ahuja et al.

    Traumatic spinal cord injury-repair and regeneration

    Neurosurgery

    (2017)
  • C.S. Ahuja et al.

    The leading edge: Emerging neuroprotective and neuroregenerative cell-based therapies for spinal cord injury

    Stem Cells Translational Med.

    (2020)
  • V.E. Arana-Argáez et al.

    Anti-inflammatory effects of Chrysophyllum cainito fruit extract in lipopolysaccharide-stimulated mouse peritoneal macrophages

    Inflammopharmacology

    (2021)
  • M.S. Beattie et al.

    Review of current evidence for apoptosis after spinal cord injury

    J. Neurotrauma

    (2000)
  • H. Cao et al.

    Liposomes coated with isolated macrophage membrane can target lung metastasis of breast cancer

    ACS Nano

    (2016)
  • Z. Chai et al.

    Ligand-Modified cell membrane enables the targeted delivery of drug nanocrystals to glioma

    ACS Nano

    (2019)
  • V. Cheung et al.

    Methylprednisolone in the management of spinal cord injuries: Lessons from randomized, controlled trials

    Surg. Neurol. Int.

    (2015)
  • F.C. da Silva et al.

    Sensory and motor responses after photobiomodulation associated with physiotherapy in patients with incomplete spinal cord injury: clinical, randomized trial

    Lasers Med. Sci.

    (2020)
  • S. David et al.

    Repertoire of microglial and macrophage responses after spinal cord injury

    Nat. Rev. Neurosci.

    (2011)
  • D.T. Ersayli et al.

    Effects of perioperatively administered bupivacaine and bupivacaine-methylprednisolone on pain after lumbar discectomy

    Spine

    (2006)
  • G. Gao et al.

    N-terminal cleavage of bax by calpain generates a potent proapoptotic 18-kDa fragment that promotes bcl-2-independent cytochrome C release and apoptotic cell death

    J. Cell. Biochem.

    (2000)
  • S.J. Gao et al.

    New approach to treating spinal cord injury using PEG-TAT-modified, cyclosporine-A-loaded PLGA/polymeric liposomes

    J. Drug Target.

    (2017)
  • Cited by (4)

    View full text