Hypoxic ischemic brain injury causes permanent damage to movement, sensation, perception, cognition, communication, behavior, and musculoskeletal problems in children. The effective treatment to recover the HIBD remains open. The mild hypothermia therapy provides neuroprotection in children with hypoxic-ischemic encephalopathy, but the improvement in prognosis is not significant.
Recent studies demonstrates that hUC-MSCs therapy was effective on treatment of HIBD. Mesenchymal stem cells have low immunogenicity to be transplanted through autologous or allogeneic transplantation applications for treatments of a variety of diseases(Weng et al. 2022). For example, MSC performed as a potential treatment to prevent complications in association with acute graft-versus-host disease (GVHD) after hematopoietic stem cell transplantation (Brooks et al. 2022). Due to the safety of MSCs, we did not use immunosuppressants in this study.
Previous studies showed that the mechanisms of MSCs’ neuroprotective effect on hypoxic-ischemic injury included inhibiting apoptosis of nerve cells, migrating to the site of brain injury, reducing the volume of infarction, and inhibiting secretion of inflammatory factors(Li et al. 2020b). Many animal experiments also showed that MSCs transplantation can restore motor function and improve memory in rats. A study(Zhang et al. 2019) showed that transplantation of MSCs after hypoxia ischemia can reduce the number of astrocytes, the loss of striatal neurons and significantly improving brain function in rats during recovery. It has been shown that MSCs can migrate to the infarct site of hypoxic-ischemic brain injury, differentiate into neurons or Schwann cells through paracrine effects, and promote nerve regeneration to function(Cai et al. 2017; Li et al. 2020b; Heris et al. 2022). The beneficial effect of stem cell therapy for brain injury models in rat function recovery and survival had been demonstrated in various animal studies(Sun and Kurtzberg 2021; Muhammad et al. 2022). Neuroprotection, neovascularization, neuronal regeneration, microglial pyroptosis and ferroptosis had all been proved after cell transplantation in animal models(van Velthoven et al. 2010; Xing et al. 2014; Huang and Zhang 2019; Hu et al. 2021; Zhai et al. 2022).
At present, stem cell therapy for cerebral ischemia and hypoxia has been applied in clinical trials and has shown good efficacy(Luo et al. 2023). However, the best practice of hUC-MSCs therapy remains open. The controversial issues such as optimal cell type, dose, timing, route of administration, and other variables still need to be refined for clinical treatment.
In our study, we investigated the best dose of hUC-MSCs for HIBD treatment by comparing four doses, 0.5* 106 (MSC1), 1.0* 106 (MSC2), 1.5* 106 (MSC3), and 2* 106 (MSC4) group.
In an earlier study(Tanaka et al. 2018), two different doses of HUC-MSCs in neonatal stroke mice showed that the high-dose(1*105) HUC-MSC treatment was significantly decreased compared with the vehicle treatment on IBA1 positive staining area of cortex. In addition, Donega et al. 2013 have explored the adequacy of different doses of stem cells in the cure of cerebral palsy rat. The study used three different doses of stem cells (0.25*106, 0.5*106, 1*106) to inject through the nose. They showed that both the 0.5*106 and 1*106 MSCs groups significantly improved the motor function of rats, while the low-dose 0.25*106 group had no significant effect, and increasing the dose to 1.0×106 did not further improve sensorimotor function or reduce cerebral infarction area. Therefore, they concluded that at least 0.5*106 mesenchymal stem cells are required in the HI-induced brain injury model to have a long-term effect on functional recovery and infarct size.
Intranasal administration is non-invasive thus can protect rat from invasive risks, and previous studies showed that intranasal injection is safe and effective, we chose the nasal route. The success of MSC transplantation is also affected by the time of transplantation. The blood-brain barrier (BBB) may lead to the failure of stem cell transplantation. Hypoxic-ischemic injury can cause the opening of the blood-brain barrier, which consecutively leads to a series of secondary pathologies. Early BBB opening occurs within hours of ischemia and may close by itself(Ek et al. 2015; Hatayama et al. 2022; Park et al. 2023). The second stage of BBB begins 24–72 hours after ischemic injury(Lehner et al. 2011; Abbasloo et al. 2023). In short, early BBB correspondence is damage to tight junction function; while late BBB opening may be associated with neuroinflammation and disruption of function(Ek et al. 2015; Park et al. 2023). The mechanisms of BBB include recruiting immune cells(Mei et al. 2021), regulating metalloproteinases(Lan et al. 2019), maintaining morphological stability(Gussenhoven et al. 2019), and the interaction of various cellular components of the blood-brain barrier(Tjakra et al. 2019). As MSCs can only reach the site of injury for a short time, transplantation too early or too late may not be effective. We used the 72-hour time point in our study.
Although there are many experiments that have proved that mesenchymal stem cells are effective in treating rats with hypoxic-ischemic brain injury. However, few studies have been able to compare the effects of treatments between different dose groups of stem cells. Therefore, we designed behavioral experiments and brain pathology sections to assess motor and learning capacity, as well as pathological manifestations. In addition, our study confirmed the efficacy of four different doses of mesenchymal stem cells in rats with hypoxic-ischemic brain injury.
The Morris Water Maze test showed that the MSC3 (1.5*106) group performed best, with the shortest time to find the platform, suggesting that it was the best on learning and memory function. In addition, the hanging wire test compared the motor function of rats’ forearm muscles, which also showed MSC3 performed best on recovering motor function.
Inflammation is one of the main pathogenic manifestations of hypoxic-ischemic brain injury, and microglia mainly activate the inflammatory response(Li et al. 2020a). Microglia are widely distributed in the brain and acted as an immune role in injury(Li et al. 2020a; Shao et al. 2021). In response to various stimuli, such as inflammation, hypoxia, ischemia and tumors, peroxiredoxin released extracellular activates Toll-like receptors and induces the production of inflammatory cytokines such as Interleukin-1β, IL-23, and Tumor necrosis factor α(Mika et al. 2013; Hickman et al. 2018; Donnelly et al. 2020). Moreover, it has been shown that microglia are more prevalent in the cerebral cortex and hippocampus than in the midbrain and hypothalamus(Fleiss et al. 2021). We thus chose the cortex and hippocampus to compare the microglia activation in different groups. The widely used microglial marker, ionized calcium-binding adapter molecule 1 (IBA1), was also used for comparison of neuropathologically-standard hypoxic ischemic injury. It showed that the IBA1-positive region of all MSC groups (MSC3 group performed best) were significantly smaller than the HIBD group in hippocampus CA1, CA3 and cortex. This suggests that the underlying mechanism of mesenchymal stem cell therapy for hypoxic-ischemic brain injury may be related to reducing the microglial activation.
Astrocytes are the largest class of glial cells in the mammalian brain and play an important neuroprotective role in cerebral ischemic injury (Jiao et al. 2020). A well-known astrocytes marker, GFAP was used to detect the morphological changes and number of astrocytes (Hol and Pekny 2015). It has been shown that the reduction of early astrocytes was closely related to clinical symptoms(Colombo and Farina 2016); in chronic central nervous system diseases, astrocytes can produce lactose ceramide, promote inflammatory response and lead to neurodegeneration(Mayo et al. 2014). These results suggest that astrocytes are two-sided effects, depending on the timing of action, specific diseases, and different stimuli of the microenvironment. Our results showed that astrocyte activation (more GFAP positive expression) was truly boosted in the HIBD group compared with the sham group, and all the MSC groups performed comparably on reducing the astrocyte activation of HIBD. It also indicated that the mechanism of MSC in the treatment of cerebral ischemia and hypoxia may be closely associated with the activation of astrocytes.
However, there are still some limitations in our study. First, although we use the well-known modified mice method for hypoxic-ischemic brain damage, this method, like all animal models, does not fully simulate human disease. Alternative methods, such as bilateral common carotid artery occlusion, are known to produce significant hypoxic-ischemic brain injury in both location and magnitude. Therefore, it is necessary to validate the results of our study using other types of HIBD animal models in newborn rats. Secondly, to achieve the best clinical treatment efficacy, problems such as route of administration, timing of administration, injection rate, and transplantation frequency still need to be addressed. Therefore, there is a long way to go to translating animal experiments into clinical treatments.