Next Article in Journal
Comprehensive Analysis and Characterization of the GATA Gene Family, with Emphasis on the GATA6 Transcription Factor in Poplar
Next Article in Special Issue
Cultured Mesenchymal Cells from Nasal Turbinate as a Cellular Model of the Neurodevelopmental Component of Schizophrenia Etiology
Previous Article in Journal
The P2X7 Receptor in Autoimmunity
Previous Article in Special Issue
Adipose-Derived Stem Cells Spontaneously Express Neural Markers When Grown in a PEG-Based 3D Matrix
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Novel Therapeutic Opportunities for Neurodegenerative Diseases with Mesenchymal Stem Cells: The Focus on Modulating the Blood-Brain Barrier

by
Pablo Vargas-Rodríguez
1,†,
Alejandro Cuenca-Martagón
2,†,
Julia Castillo-González
1,
Ignacio Serrano-Martínez
1,
Raúl M. Luque
2,3,4,5,
Mario Delgado
1 and
Elena González-Rey
1,*
1
Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016 Granada, Spain
2
Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain
3
Department of Cell Biology, Physiology, and Immunology, University of Cordoba, 14004 Cordoba, Spain
4
Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain
5
CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(18), 14117; https://doi.org/10.3390/ijms241814117
Submission received: 2 August 2023 / Revised: 8 September 2023 / Accepted: 12 September 2023 / Published: 14 September 2023
(This article belongs to the Special Issue Mesenchymal Stem Cells in Neurological Disorder)

Abstract

:
Neurodegenerative disorders encompass a broad spectrum of profoundly disabling situations that impact millions of individuals globally. While their underlying causes and pathophysiology display considerable diversity and remain incompletely understood, a mounting body of evidence indicates that the disruption of blood-brain barrier (BBB) permeability, resulting in brain damage and neuroinflammation, is a common feature among them. Consequently, targeting the BBB has emerged as an innovative therapeutic strategy for addressing neurological disorders. Within this review, we not only explore the neuroprotective, neurotrophic, and immunomodulatory benefits of mesenchymal stem cells (MSCs) in combating neurodegeneration but also delve into their recent role in modulating the BBB. We will investigate the cellular and molecular mechanisms through which MSC treatment impacts primary age-related neurological conditions like Alzheimer’s disease, Parkinson’s disease, and stroke, as well as immune-mediated diseases such as multiple sclerosis. Our focus will center on how MSCs participate in the modulation of cell transporters, matrix remodeling, stabilization of cell-junction components, and restoration of BBB network integrity in these pathological contexts.

1. Introduction

The evidence indicates that longevity is currently increasing worldwide, and the occurrence of cumulative disabilities is the price to pay for living longer [1]. It is widely recognized that aging is a natural, progressive, and inevitable process that occurs in all organisms, although the functional and morphological changes affecting the body tissues and organs during its progression are highly variable [2]. Specifically, aging-associated changes in the central nervous system (CNS) are of crucial relevance, and the prevention and treatment of neurodegenerative conditions represent one of the greatest challenges for modern societies. However, therapeutic options to treat CNS-related disorders are very limited, mainly due to the fine-tuned status of the brain, the complexity of neurological diseases, and the lack of knowledge of their etiology and pathophysiology.
An additional challenge in this scenario comes from the singular presence in the CNS of the blood-brain barrier (BBB), a complex, dynamic, and structured network of cells and proteins responsible for protecting the brain and regulating the transport of substances and cells from the peripheral circulation to the CNS. Because keeping the integrity of the BBB is critical for maintaining a constant environment in the CNS in healthy conditions, investigating how this structure changes during aging, and specifically in pathological conditions, has lately received special attention worldwide. Although some researchers have demonstrated minor and variable BBB leakage in healthy aging without immune infiltration and neuropathological signs [3], evidence indicates that BBB integrity is compromised in most neurological disorders, including those associated with aging [4,5,6,7,8,9,10,11]. Therefore, BBB disruption emerges as a key innovative element to design new therapeutic approaches to treat neurodegeneration during aging and neurological afflictions.
Recently, stem cells isolated from adult mesenchymal tissues (MSCs) have emerged as attractive candidates for the treatment of aging-associated neurological diseases [12,13]. While many studies have explored the involvement of the neuroprotective, neurotrophic, and immunomodulatory capacities of MSCs in their therapeutic actions in neuroinflammatory and neurodegenerative disorders, the potential additional role played by MSCs in improving the sealing and modulation of the BBB has scarcely been addressed.
This review aims to examine the structure and functions of the BBB and how its impairment, along with changes in transporters, extracellular matrix, and cell-junctional components, influences the onset and progression of several neurodegenerative disorders. We will specifically focus on four neurodegenerative conditions, namely Alzheimer’s disease (AD), Parkinson´s disease (PD), multiple sclerosis (MS), and stroke. While AD, PD, and MS are chronic diseases with etiologies that remain mostly unknown, stroke is mainly recognized as an acute pathological state of the brain with well-described pathogenesis and etiology. Although these disorders exhibit different clinical and pathological signs, they all progress with neuroinflammation and neurodegeneration, are significantly associated with aging, and display profound alterations in cerebral vasculature and microvessel components. Importantly, they all respond positively to MSC-based therapies. Our review will also delve into the molecular mechanisms involved in the treatment of neurological pathologies with MSCs, with special attention to their ability to restore the integrity of the BBB. Finally, we will discuss how to further improve MSC therapies by specifically targeting the BBB.

2. Structure and Role of the Blood-Brain Barrier

The BBB is a complex and dynamic microvascular structure composed of several types of cells, which maintains the homeostasis of the CNS by regulating the supply of molecules and filtering potentially harmful compounds from the bloodstream to brain tissues and back [14]. Its main scaffold is structured into a neurovascular unit (NVU) that is shaped by a tight layer of brain microvascular endothelial cells (BMVECs) surrounded by astrocyte end-feet and pericytes, all of them embedded in an extracellular matrix network and the basement membrane [15] (Figure 1).
The BMVECs are highly specialized endothelial cells that show unique structural and biological properties compared to peripheral endothelial cells [16], including low pinocytic activity, absence of fenestration, low levels of leukocyte adhesion molecules, high expression of intercellular junctions, mainly tight junctions (TJs) and adherens junctions (AJs) [17,18], and increased number of mitochondria to supply the energy that is required for the active transendothelial transport of molecules [19]. Additionally, BMVECs regulate BBB permeability by mainly controlling the intercellular and intracellular transport of cells and molecules via cellular junctions and specific membrane carriers, channels, and transporters [20]. Moreover, these cells present a very remarkable apicobasal polarity, based on a differential membrane composition (lipids, glycoproteins, receptors, and transporters) between the luminal and the abluminal sides [21]. For instance, enzymes like γ-glutamyl-transpeptidase [22] or alkaline phosphatases [23] are located in the luminal face of the endothelium, while Na+-K+ ATPase [24] and the Na+-dependent neutral amino acid transporter [25] are present at the basal membrane.
BMVECs are wrapped by pericytes which contribute to the regulation of endothelial cell proliferation, survival, migration, differentiation, vascular branching, and blood flow control [26]. Smooth muscle cells are also found around large vessels (arteries, arterioles, venules, and veins), providing strength and elasticity, and playing a significant role in basal tone maintenance, blood pressure, and blood flow distribution [27,28]. Pericytes are localized along capillaries and embedded within the BM. They regulate blood flow, modulate immune and phagocytic responses after brain injury, and promote angiogenesis in the adult CNS [29,30].
Astrocytes are the major glial cell that enfolds the endothelium of the BBB [31]. They connect to BMVECs through their end-feet, contributing to BBB consistency and determining its properties [30]. Astrocytic end-feet contain a set of proteins that interact with the vascular tube, such as dystroglycan-dystrophin complex or aquaporin 4 (AQP-4), among others. The latter is critical for regulating water homeostasis in the CNS, while the dystroglycan-dystrophin complex links the astrocytic skeleton to the BM [32]. Since astrocytes serve as the cellular linkage between the neuronal circuitry and the vascular system in the CNS, they release signals that regulate the blood flow in response to neuronal activity. For instance, they control the contraction and dilation of SMCs and pericytes [33]. Additionally, they contribute to the formation of endothelial cell TJs through vascular endothelial growth factor (VEGF)-mediated signals [34] and regulate tissue inhibitor metalloproteinases (TIMPs), which maintain the balance between deposition and degradation of the extracellular matrix components [35].
The basement membrane (BM) is a highly organized sheet composed prominently of extracellular matrix proteins (collagen IV, laminins, nidogen, and perlecan). It plays an important role in providing structural support, cell anchoring, and signaling transduction [36]. Two types of BM of the BBB have been characterized: the inner vascular BM, secreted by BMVECs and pericytes, which contains laminins α1 and α2; and the outer parenchymal BM, secreted by astrocytes, which contains laminins α4 and α5 [37,38]. Nevertheless, the BM is largely understudied, in comparison to the cellular components of the BBB, probably due to its intrinsic complexity and the lack of research tools [39].
The exchange of molecules and cells across the BBB requires specific transporters, channels, and receptors. Two major mechanisms, named paracellular and transcellular transport, have been identified. Table 1 describes some of the main transporters. Paracellular transport is the main pathway used for the exchange of small hydrophilic substances. On the other hand, the transcellular transport involves the movement of molecules through the cell membrane of BMVECs. It occurs via several mechanisms depending on the nature of the molecule, such as passive diffusion, facilitated diffusion, active transport, and receptor-mediated transport. Importantly, gases and small lipophilic molecules do not require transporters to cross the BBB and freely diffuse across the endothelium [40].
Both paracellular and transcellular processes are precisely regulated by junctional structures, mainly TJs, with some contribution from gap junctions and AJs [41]. TJs are intricate structures located along the membranes of adjacent BMVECs, intermingled with AJs, providing stability and consistency to the BBB [42,43] (Figure 1). Among TJ proteins, claudin-1, -3, -5, -12, and occludin control the transportation of solutes and ions [44]. These proteins are associated with the cytoskeleton, primarily based on actin and vinculin, through scaffolding proteins, such as ZO-1, -2, and -3. Moreover, dystrophin acts as a scaffold protein that mobilizes actin and vinculin proteins [45]. AJs create inter-endothelial contact connections, maintained by proteins such as VE-cadherin and platelet endothelial cell adhesion molecule-1 [46,47] (Figure 1), that contribute to the continuous crosstalk with TJs for paracellular transportation. Similarly to TJs, AJs are attached to the cytoskeleton, contributing to the regulation of cellular transportation of lymphocytes, monocytes, or neutrophils [48,49,50]. Pericyte-endothelial junctions also contain cadherins, with N-cadherins forming homophilic interactions between pericytes and BMVECs, thereby maintaining vascular integrity [51]. Furthermore, BMVECs interact with the BM, establishing AJs via α- and β-integrin receptors, which are transmembrane glycoproteins involved in the extracellular matrix connection to the endothelial cytoskeleton [44]. Conversely, gap junctions serve as intercellular channels facilitating cytoplasmic connections between neighboring cells, enabling selective communication of molecules primarily dependent on molecular size, driven by passive diffusion [52]. In the brain, endothelial cells express the gap junctions connexin 37 (Cx37), Cx40, and Cx43 [53], while astrocytes express Cx30 and Cx43 [54]. In addition to their channel function, certain connexins also play a regulatory role in the expression of other junctional molecules, such as Cx43 interacting with N-cadherin [55].
Table 1. Proteins involved in transcellular transportation in BMVECs of the BBB.
Table 1. Proteins involved in transcellular transportation in BMVECs of the BBB.
TransporterCargoLocationDescriptionSource
Glucose Transporter 1
(GLUT-1)
GlucoseAbluminal and luminal sideMain glucose transporter of BMVECs. Also expressed in astrocytes but not in neurons. Na+ dependent transporters[40,56]
Large neutral amino acid transporter 1
(LAT1)
Large neutral amino acidsAbluminal and luminal sideAbluminal side LAT1 transport is dependent of Na+ concentration. Bidirectional transport[40,57]
Cationic amino acid transporter 1 and 3 (CAT1/3)Cationic amino acidsAbluminal and luminal sideCAT-1 is pH and Na+ independent but sensitive to changes in membrane potential[58]
Na+-dependent transporters for glutamate exist on astrocytes 1 and 2 (EAAT1/2)GlutamateAbluminal sideExpressed in astrocytes. Possible protective mechanism against glutamate neurotoxicity[59]
Monocarboxylate transporters (MCT1)Monocarboxylic acids (lactate, pyruvate, and acetoacetate and β-hydroxybutyrate)Abluminal and luminal sideIntracerebral transport. Located in BMVECs and astrocytes. The transport mechanism is a H+ cotransporter or a monocarboxylate exchanger[60,61]
Insulin receptor (IR)InsulinAbluminal and luminalLocated in BMVECs. Insulin binding activates IR by phosphorylation of beta-chain region. Impaired phosphorylation response in AD[62]
Low-density lipoprotein receptor–related protein 1 (LRP1)APO2 and APO3Mainly in the abluminal sideLocated in BMVECs. LRP1 binds to Aβ aggregates and mediates their clearance from brain to blood. LRP1 level diminished in AD patients leads to aggregates accumulation[26,63]
Receptors for advanced glycation end-products (RAGE)Advanced glycation end products (AGE), high mobility group box-1 (HMGB-1) proteinMainly at the luminal sideLocated in BMVECs, microglia, and astrocytes. Upregulated in AD. It mediates the influx of Aβ into the brain[64,65]
P-glycoprotein, ATP-binding cassette 1
(P-gp, ABCB1)
Xenobiotics and drugsExpressed in the luminal sideP-gp is a unilateral efflux pump from blood to brain. It uses ATP in the active transport of substances. It is crucial in the ADMET properties of pharmaceutical drugs. In AD, P-gp is involved in accumulation of Aβ peptides in the CNS[66,67]
Transferrin receptor protein (Tfr)Transferrin (apo- and holo-transferrin)Abluminal and luminal sidePrimary iron transporting system. Highly enriched in BMVECs. Studied as a targeted transporter of therapeutics to the brain[68]

3. MSCs as a Therapeutic Option in CNS Disorders

Despite notable advancements in the management of symptoms accompanying the neurodegeneration/neuroinflammation in AD, PD, MS, and in the development and progression of acute stroke, with treatments that enhance quality of life and increase lifespan, the available drugs only slow the progression of neuronal death. Given the multifactorial and complex nature of these conditions, the primary causal agent remains unclear, and it is imperative to develop multi-target therapies that address the different causes/consequences of these disorders, such as neuroinflammation, neuronal cell death and dysfunction, and BBB disruption.
MSCs are emerging as one of the most promising cell therapies against different immune-mediated diseases due to their unique properties. MSCs are multipotent cells able to differentiate into mesodermal lineages (fibroblast, osteocyte, adipocyte, and chondrocyte) and, in some cases, into endodermal or ectodermal (neuronal) fates [69]. The scarce expression of the major histocompatibility complex and other co-stimulatory molecules makes MSCs immune-privileged cells. This immune status allows MSCs to be used in an allogenic manner without requiring additional immunosuppression [70].
The International Society for Cellular Therapy has defined MSCs based on their expression of CD90, CD73, CD105, and CD44, while lacking the expression of CD45 and CD31 [71]. These markers help to distinguish MSCs from other cell types and are used to identify and isolate these cells for research and therapeutic purposes.
In adults, several tissues act as MSC reservoirs [72]. The first type of MSC to be described were bone marrow-derived mesenchymal stem cells (BM-MSCs) [73], making BM the primary source for MSC isolation. However, the process of obtaining BM-MSCs involves a highly invasive and painful procedure that requires anesthesia, posing a risk of infection [74]. Alternatively, adipose tissue-derived MSCs (ASCs) can be isolated from biological material generated during liposuction or lipectomy after medical interventions. The natural abundance of MSCs in adipose tissue, which is approximately 500 times higher than in BM, accompanied by easier isolation, has led to an increased utilization of ASCs [75]. Additionally, a recent study has demonstrated that ASCs exhibit lower immunogenicity and transcriptomic heterogeneity compared to BM-MSCs [76]. Apart from adult tissues, MSCs can also be derived from birth-associated tissues, such as the umbilical cord Wharton’s jelly (WJ). WJ-MSCs have emerged as an ideal source of MSCs for therapy due to several advantages: they can be harvested painlessly in abundance without causing donor site morbidity, are easy to isolate and culture, possess a high proliferative rate, and retain their stemness properties in vitro [77].
In the context of neurodegeneration, there is growing interest and promise in therapies based on MSCs. As of July 2023, 249 clinical studies were found on www.clinicalstrials.gov [78] with the terms “Nervous System Diseases” and “Mesenchymal Stem Cell”, reflecting the potential of these cells in addressing neurodegenerative/neuroinflammatory disorders. A summary of studies focused on AD, PD, MS, and stroke (a total of 96 from the 249 trials) identifying the tissue for MSC isolation, donor, route of administration, and pathological target is shown in Table 2.
The main mechanisms exerted by MSCs that contribute to their potential efficacy include:
  • Neuroprotective effect: MSCs have demonstrated to have an important neuroprotective effect, as they secrete neurotrophic growth factors such as glial cell-derived neurotrophic factor, VEGF, brain-derived neurotrophic factor, and nerve growth factor (NGF) [79], as well as anti-apoptotic factors like Bcl-2 [80]. These factors enable MSCs to promote nervous regeneration, inhibit neuronal apoptosis, and induce endogenous neurogenesis. For example, Oh et al. [81] demonstrated that intravenous injection of MSCs increased hippocampal neurogenesis and differentiation of neural progenitor cells into mature neurons in Aβ-treated mice (AD model) by augmenting the Wnt signaling pathway. Additionally, MSCs may inhibit stroke-associated apoptosis through the Bcl-2 pathway in neurons and astrocytes from rats [82]. Furthermore, MSCs can transfer healthy mitochondria to damaged cells, protecting neural stem cells from neurotoxic agents. MSCs may transfer this organelle in various ways, including gap junctions, cell fusion, microvesicles, and through tunnelling nanotube formation [83]. Mitochondria play a crucial role in maintaining metabolic homeostasis, and defects such as membrane leakage, electrolyte imbalances, activation of pro-apoptotic pathways, and mitophagy have been implicated in the pathogenesis of various CNS disorders [84]. It has been demonstrated that the ability of MSCs to transfer healthy mitochondria to damaged cells protects neural stem cells from neurotoxic agents [85], and has garnered significant attention in the field of cellular therapy for CNS disorders;
  • Immunomodulatory role: MSCs can interact with the immune system and participate in both innate and adaptive immunity due to their significant immunoregulatory functions. This indicates that, depending on the environment in which MSCs are introduced, they can modulate the response. Thus, in an inflammatory environment, MSCs exhibit anti-inflammatory behavior. By expressing different molecules such as transforming growth factor β, indoleamine 2,3-dioxygenase, prostaglandin E2, nitric oxide, and interleukin-10 (IL-10), they can interact with immune cells either through direct cell-to-cell contact or via paracrine activity [86,87,88,89,90]. MSCs can also modulate the macrophage/microglia polarization, upregulating the ratio of anti- versus pro-inflammatory responses [91], suppress Th1 and Th17 responses, enhance the maturation of DCs from monocytes, and enhance the Th2 response and the generation of Forkhead Box P3 positive Treg population. Moreover, some studies reported that the secretion of IL-6 by MSCs can inhibit astrocyte apoptosis, increase the neuroprotective population of astrocytes, and reduce neuron damage post-injury [92];
  • Regulation of protein clearance: treatment with MSCs has been shown to induce the secretion of neprilysin in vitro and in vivo, improving the endogenous machinery for the degradation of Aβ-plaques and enhancing the clearance of these aggregates [93]. This is particularly relevant as abnormal protein aggregation is one of the major hallmarks of neurodegenerative diseases like PD and AD [94].

4. MSCs as Promising Modulators of the BBB in Neurodegenerative Disorders

As described before, while it remains unclear whether BBB disruption is a cause or a consequence of neuroinflammation, it is undoubtedly a crucial component of CNS pathologies. Unfortunately, the BBB is often viewed as a challenge that hinders the delivery of drugs to the CNS and reduces the efficacy of conventional treatment approaches for neurodegeneration. Therefore, the potential of pharmacological interventions targeting the BBB could represent a promising therapeutic strategy for the neuroinflammatory-mediated neurodegenerative diseases [12,13]. In addition to the neuroprotective and immunomodulatory roles of MSCs in neurodegeneration, recent reports have pointed out a beneficial effect of MSCs on modulating the disrupted BBB.
In general, the delivery of MSCs to the CNS is highly diverse, although systemic administration, particularly intravenous infusion, is the preferred method (Table 2). When MSCs are infused intravenously, they transiently accumulate in the lungs for 1–3 h, followed by a gradual movement to other tissues such as the liver, spleen, kidney, and bone marrow [95]. Interestingly, MSCs have shown the ability to reach brain vessels and adhere to them 6 h post-injection, according to Rüster et al. [96], thanks to specific interactions with endothelial cells through adhesion molecules such as P-selectin and VCAM-1/VLA-4. While some reports indicate that, after a middle cerebral artery occlusion (MCAO) model of stroke, injected MSCs accumulate in the vessels of the infarcted region [97,98], other studies describe no MSCs being detected in the cerebral parenchyma after an intra-arterial injection in an Alzheimer’s disease mouse model [99]. Therefore, although MSCs are capable of rolling and executing a coordinated extravasation through activated endothelia in other tissues, allowing them to access sites of damage [96], it remains unclear whether this cellular therapy can cross the BBB and exert its function within the brain tissue.
Nevertheless, despite their uncertain ability to cross the BBB and penetrate to the brain, MSCs can directly make contact with endothelial cells in the damaged area. This interaction likely enables MSCs to exert their paracrine functions to other cells of the NVU and the BBB from this location (Figure 1). In fact, the scientific community is currently exploring two interesting derivatives of MSCs: genetically modified MSCs and the use of MSC-derived extracellular vesicles (MSC-EVs). Genetically modified MSCs provide the opportunity to enhance the therapeutic effect of MSCs by improving their inherent functions or enabling them to synthesize drugs or active compounds. On the other hand, direct use of the secretome in the form of MSC-EVs improves penetration through the BBB. In the following sections, we will provide examples of these strategies and the cellular and molecular beneficial effects exerted by MSCs in preclinical models of neurodegenerative conditions characterized by a severe disruption of the BBB.

4.1. Alzheimer’s Disease

Alzheimer´s disease (AD) is a progressive neurodegenerative disease characterized by a cerebrovascular and neuronal dysfunction, resulting in a gradual decrease in cognitive functions [100]. In 2019, it was estimated that 50 million people suffered from AD [101]. The principal pathological hallmarks are extracellular amyloid-β (Aβ) deposition and neuronal accretion of phosphorylated tau-forming neurofibrillary tangles [102]. Aβ is a proteolytic by-product derived from the amyloid precursor protein, produced by several cleavages via β- and γ-secretases. There are two main types of AD: early onset AD, which is a rare form affecting <1% of AD cases in subjects <65 years old and is caused by genetic mutations, and late onset AD, which is the most frequent form, and primarily affects to patients >65 years old. While genetic factors may potentially contribute to its development, specific mutations that directly cause an increase in proteolytic cleavage in patients have not been observed [103].

4.1.1. Dysfunctional BBB in AD

BBB dysregulation plays a significant role in the pathogenesis of AD, affecting various components of the NVU (Figure 2).
For instance, the presence of Aβ disrupts the organization of TJs and AJs (i.e., occludin, claudin-5, and ZO-1) in BMVECs, leading to compromised barrier activity [104,105]. Additionally, the reduction in GLUT-1 in cerebral microvessels of AD patients contributes to vessel degeneration and further exacerbates the disease [106,107]. Conversely, patients with mild cognitive impairment, which is the precursor of AD, display increased BBB permeability that correlates with high levels of soluble platelet-derived growth factor receptor β in the cerebrospinal fluid, which is indicative of pericyte damage [108]. In fact, the decreased number of pericytes in AD patients may worsen the accumulation of Aβ both in brain parenchyma and blood vessels.
On the other hand, astrocytes in AD patients showed reduced expression of AQP4 in perivascular end-feet and increased levels of astrocytic activation markers [109]. In fact, the accumulation of Aβ in the brain leads to pericyte degeneration and loss, a dysregulated BM, and astrocytic end-feet depolarization with loss of AQP4, which will decrease the Aβ clearance, fueling a pathogenic feedback loop. Thickening of the BM [110] and increased collagen levels in these structures [111,112] are also common features in AD patients. MMP2 and MMP9 are significantly activated in the NVU, contributing to BM remodeling in AD [113,114].
Furthermore, transporters of the BBB, such as RAGE, LRP1, or P-gp, are key elements in the regulation of Aβ clearance. In fact, analysis of microvessels and BMVECs in postmortem AD brains showed high expression of RAGE, which mediates Aβ entry into the brain [115], and reduced expression of LRP1 and P-gp, involved in the clearance of cerebral Aβ [116,117,118]. The immune system is also compromised in AD, and cells like monocytes, lymphocytes, or neutrophils can cross the BBB in response to Aβ accumulation and the augmentation of vascular adhesion molecules, contributing to the pathogenesis of AD [119,120,121].

4.1.2. Therapeutic Opportunities for MSCs Targeting the BBB in AD

Currently there are no effective treatments to cure or slow AD progression. However, emerging evidence suggest that MSC therapy could be a promising approach. In general, MSC transplantation has been found to decrease Aβ deposits and plaques, and tau-related cell death in vivo. The paracrine effects of MSCs stimulate neurogenesis, synaptogenesis, and neuronal differentiation, demonstrating neuroprotective functions. Moreover, their immunoregulatory properties, which modulate microglia/astrocytes’ activity state, can deactivate neuroinflammatory responses via several transcription factor signaling pathways [122].
The BBB represents a major challenge in treating AD, and different studies have focused on the action of MSCs on cerebral vasculature (Figure 2). For instance, Garcia et al. [123] demonstrated the ability of intracerebrally transplanted MSCs in a 2xTg-AD mouse model to promote neovascularization in the hippocampus. Specifically, they found that MSCs genetically modified to express VEGF enhanced their therapeutic efficacy in promoting neovascularization. Focusing on transporters involved in AD, Son et al. [124] modified MSCs to express the secreted isoform of RAGE (sRAGE), which inhibits the interaction between RAGE and its ligands, thus preventing the adverse effects of this signaling pathway. When activated by Aβ oligomers, RAGE can lead to cell stress, generation of ROS, and RAGE-mediated inflammation and neurodegeneration. Transplantation of sRAGE-MSCs into 5xFAD transgenic mice reduced the deposition of Aβ, cell death, and inflammation.
In a rat model of cerebral small vessel disease, a pathology characterized by Aβ deposition equivalent to AD, the intravenous infusion of MSCs restored the polarity/distribution of AQP4 to the end-feet of astrocytes, relieving cerebral edema and promoting the clearance of Aβ [125]. In another study, Tachibana et al. [126] implanted mouse MSC-derived pericytes into the brains of APP/PS1 mice and observed a reduction in Aβ levels in the hippocampus, an effect that was mediated by LRP1. Interestingly, a recent study has shown that, in a model of microfluidic BBB-like microvasculature, BM-MSCs emulate more efficiently the function of perivascular pericytes than induced pluripotent stem cell-derived pericytes, leading to greater restoration of TJs and the abluminal BMs [127]. In fact, there are several similarities between MSCs and pericytes. Pericytes express a similar pattern of immunological markers (CD44, CD90, CD73, CD105, and CD45), are self-renewable, and have the capacity to differentiate into nervous cells, mainly glial cells, in vivo [128]. Therefore, MSCs could potentially supply the loss of pericytes in AD. In summary, these results highlight the need for further research in this field, as understanding the role of MSCs in modulating the BBB in the context of AD is essential to develop effective therapies.

4.2. Parkinson’s Disease

Parkinson´s disease (PD) is the second most common neurodegenerative disease after AD. This progressive disorder is characterized by the loss of dopamine neurons in the substantia nigra pars compacta (SNpc) and the accumulation of filamentous and oligomeric inclusion bodies (Lewy bodies) composed of misfolded α-synuclein proteins. These structures disrupt cellular processes and lead to neuronal degeneration, mainly leading to motor dysfunction. Its etiology is still unknown and current treatments are mainly focused on symptoms [129].

4.2.1. Dysfunctional BBB in PD

Although decades ago, NVU was not recognized as an important element of the pathogenesis of PD, it is now well-established that BBB disruption is associated with PD (Figure 3).
Nevertheless, further research is needed to completely understand the molecular mechanism of BBB disruption in this context [130]. Brain tissue from PD patients showed perivascular deposits of fibrinogen or fibrin, IgG, and hemosiderin in specific regions, indicating BBB disruption [131,132,133]. In fact, degeneration of BMVECs and TJs, as well as disorganization of the components of the BM, have been reported in PD brain tissues [132]. In addition, patients with idiopathic PD show genetic mutations that affect NVU components. For example, mutations in leucine-rich repeat kinase 2 in BMVECs lead to increased monocyte attachment in PD patients [134]. Moreover, several PD patients have mutations in the MDR1 gene, which encodes P-gp in BMVECs, resulting in reduced pump function [135].
Angiogenesis is also affected in PD. Although PD patients show augmented vascular density in the SNpc, in the proximity of neuronal damage, these new microvessels display impaired maturation processes and altered diameters [136]. While new microvascular architecture may allow the supply of nutrients and cellular debris, it also raises the risk of leakage and infiltration of toxins, drugs, and immune cells, potentially exacerbating the pathology [137,138]. Moreover, proinflammatory cytokines secreted by activated immune and glial cells (such as TNF-α, IL-1β, and IL-6) [139] can decrease the expression of ZO-1 and occludin, leading to a disruptive state of TJs and subsequently contributing to BBB breakdown [140].

4.2.2. Therapeutic Opportunities for MSCs Targeting the BBB in PD

Over the last decade, preclinical studies investigating the potential of MSCs in the context of PD have been performed (Figure 3). For instance, Chao et al. conducted an extensive study on the effect of MSCs using the preclinical model of PD induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) [141]. They found that MSCs, intraperitoneally infused 24 h after the last MPTP injection, migrated to the SNpc and efficiently rescued dopaminergic TH+ neurons. Moreover, the treatment with MSCs restored BBB integrity, as indicated by the retrieval of TJ-protein expression (claudin-1, claudin-5, and occludin).
Other studies have recently demonstrated that treatment with MSC-derived exosomes regulates genes associated with the angiogenesis of human BMVECs in vitro, resulting in increased expression of Angpt1 and Flk1, as well as the secretion of the intercellular adhesion molecule 1 (ICAM1) protein. Injecting these exosomes into an MPTP-induced PD model resulted in their homing to the injured brain and a significant recovery from the disease. Additionally, there was an increase in the expression of ICAM1 and CD31 markers in the striatum and SNpc [142]. Conversely, in an LPS injection model into the SNpc, treatment with MSCs increased the expression of P-gp in endothelial cells and restored BBB integrity [143]. The study suggested that MSCs decreased the proinflammatory activation of microglia and modulated the VEGF-A signaling through astrocytes, leading to an increase in the astrocytic end-feet density. This process stabilized the expression of TJ proteins. These results suggest the relevance of modulating the BBB in PD for developing effective therapies against this debilitating disease.

4.3. Multiple Sclerosis

Multiple sclerosis (MS) is an inflammatory neurodegenerative autoimmune disease that affects the CNS. In MS, the immune system generates a complex response against myelin sheaths that wraps nerve axons, eventually leading to inflammation, demyelization, axonal degeneration, and ultimately neuronal loss [144]. Immune cells cross the damaged BBB and release proinflammatory cytokines, such as TNF-α, IFN-γ, or IL-17, which directly attack myelinating oligodendrocytes [145] or provoke a pro-inflammatory polarization of microglia and astrocytes that finally causes oligodendrocytes loss [146].
There are different types of MS depending on the evolution of the disease: relapsing-remitting MS (RRMS) and primary/secondary progressive MS (PPMS/SPMS). RRMS is characterized by alternating periods of symptom enhancement (relapses) and partial or complete recovery of neurologic function (remissions). Conversely, progressive MS (15% of MS patients) is marked by a gradual worsening of symptoms without periods of relapses or remissions. On the contrary, SPMS follows the initial relapsing-remitting course [144].

4.3.1. Dysfunctional BBB in MS

Although the etiology of the disease is not completely understood, a vast body of evidence suggests the importance of BBB disruption in the pathology of MS (Figure 4).
Inflammation affects several components of the NVU and hampers the physiologic function of numerous transport mechanisms. BBB permeability may be an important early step that is correlated to the initiation of a CNS-specific immune response [147]. Pro-inflammatory mediators, such as IL-1β [148], IL-6 [149], TNF-α [150], and chemokine (C-C motif) ligand 2 (CCL2) [151], reduce the expression of TJ and AJ proteins in different in vitro BBB models. Specifically, the expression of Toll-like receptors, which play a significant role in modulating MS, is significantly increased in BMVECs in response to ROS and TNF-α [152,153]. The expression of other transporters, like GLUT-1 [154], LAT1 [155], or P-gp [156], is also affected by the exposure of BMVECs to inflammatory mediators. Moreover, pro-inflammatory molecules can cause pericyte detachment from BMVECs and undergo transformation into phagocytic or fibroblastic-like cells [147]. Regarding the autoimmune component of MS, activation of BMVECs with Th1 cytokines (IL-2, TNF-α, IFN-γ) modulates the BBB phenotype and stimulates the expression of endothelial cell adhesion molecules, such as ICAM-1 and VCAM-1 [157].

4.3.2. Therapeutic Opportunities for MSCs Targeting the BBB in MS

The disruption of the BBB in MS allows immune cells to infiltrate into the CNS, contributing to the pathogenesis of MS, as previously mentioned. In vitro studies using a BBB model exposed to TNF-α have shown that treatment with embryonic MSCs modulates barrier permeability, increases the expression of TJ proteins, and decreases the expression of pro-inflammatory chemokines like CCL2 and CXCL12 [158] (Figure 4). In vivo studies have also demonstrated that co-administration of MSCs expressing IFN-β along with minocycline tames the disruption of the Blood-Spinal Cord Barrier (the functional equivalent of the BBB in the spinal cord) [159]. Recent studies in the MS preclinical model (experimental autoimmune encephalomyelitis) have further shown that MSC transplantation reduced BBB disruption, as evidenced by reduced IgG leakage. Additionally, MSC transplantation led to the adequate expression of TJ-proteins occluding and ZO-1 in BMVECs and the restoration of AQP4 levels in astrocytes [160].

4.4. Stroke

Stroke refers to an acute brain injury derived from no other than a vascular cause, leading to neuronal damage and functional disability. As a complex and heterogeneous condition, it is influenced by genetic predisposition, aging, life habits, and chronic diseases. It is the second leading cause of death and third leading cause of disability worldwide. Notably, 60% of surviving patients are affected by cognitive impairment, dementia, or depression. Stroke is classified into two types: hemorrhagic and ischemic, with the latter being the most prevalent (87% of the total). Ischemic stroke is characterized by a sudden cessation of oxygen and blood supply due to thrombus blocking blood flow in the brain vasculature [161].This initiates a rapid and complex cascade of pathophysiological events at the genomic, molecular, and cellular levels that may evolve over hours to days and weeks after the onset, including energy failure, acidosis, loss of cell homeostasis, excitotoxicity, oxidative stress, activation of glial cells, inflammation, and disruption of the BBB with infiltration of leukocytes [162,163].

4.4.1. Dysfunctional BBB in Brain Ischemia

The BBB plays a significant role in the pathophysiology of ischemic stroke, and its dysfunction varies depending on the severity and duration of the ischemia. Predominantly, during human stroke, the BBB presents a continuous opening pattern with biphasic peaks distributed along four stages [164] (Figure 5).
The hyperacute stage is the first phase that evolves within the first 6 h after the ischemic onset. During this phase, the first BBB opening is documented. Due to oxygen and glucose deprivation, Na+ and Ca+ accumulate inside the cells of the NVU, including astrocytes or BMVECs, leading to cytotoxic edema [165], glutamate excitotoxicity [166], oxidative damage associated with ROS generation, and mitochondrial dysfunction [164]. Moreover, MMPs (mainly MMP2) directly degrade TJ proteins and BM components, contributing to BBB leakage [167].
The next stage corresponds the acute phase, which occurs after the first 6 h of the onset for a period of 72–96 h. The second permeability peak is observed at this stage. From this point, immune components start to participate more significantly to stroke pathophysiology. Neutrophils are the predominant peripheral immune cells in the acute post-stroke period [168]. They contribute to BBB disruption by producing excessive ROS [169], proteases (MMP9, proteinase-3, elastase) [170], and neutrophil gelatinase-associated lipocalin [171], generating neutrophil extracellular traps [172], and secreting inflammatory cytokines (IL-1β, IL-6, IL-8, TNF-α) and chemokines (CCL2, CCL3, and CCL5) [173]. Moreover, P-selectin glycoprotein ligand-1 and macrophage-1 antigen in neutrophils, and their respective receptors, P-selectin and ICAM-1, in BMVECs, are upregulated shortly after an ischemic stroke induced by IL-1β. Hence, paracellular BBB permeability is increased [174,175]. Natural killer (NK) cells are also present during this stage, producing IFN-ϒ and ROS [176]. Monocytes and cerebral immune cells, microglia, and astrocytes display a pro-inflammatory phenotype which exacerbates BBB breakdown through the secretion of proinflammatory factors (IL-1α, IL-1β, IL-6, TNF-α, IFN-γ, CCL2), MMP9, VEGF, ROS, and activation of inducible nitric oxide synthase [162]. Finally, upregulation of MMP-9 is crucial in this phase (24–48 h after onset), and it plays a dual role in the proteolytic degradation of the BM components of the BBB. Its ability to digest TJ proteins enhances the disruption of the BBB [177]. Concomitantly, vascular remodeling is induced by VEGF, leading to the mobilization of progenitor BMVECs, which implies an immature and damaged BBB during this neovascularization process [178].
The subacute phase starts one week after the stoke onset, a timepoint from which the BBB begins its recovery process. Monocytes and glial cells shift to anti-inflammatory phenotypes expressing cytokines (IL-10, IL-4, TNF-β) and neurotrophic factors that help prevent inflammation [179]. Moreover, during this phase, angiogenesis plays a vital role in restoring the blood flow and oxygen supply in ischemic tissues. Angiogenesis promotes the proliferation and migration of BMVECs and pericytes, enhances tube formation, branching, and anastomosis, all of which are modulated by the inflammatory microenvironment. VEGF, MMP9, and angiopoietin 2 (Ang-2), as well as its receptor Tie-2, are deeply involved in this process [164]. While these agents may temporarily contribute to BBB leakage, a higher degree of angiogenesis has been linked to increased survival in patients [180] and greater stability of the BBB [181].
Finally, the chronic post stroke phase starts approximately six weeks after the ischemic event, where the BBB is still disrupted but to a lesser extent compared to the previous phases. During this stage, NVU components are restored in order to seal the BBB, with an overexpression and new distribution of TJ proteins. Some of the factors that stabilize the BBB include Ang-1, which maintains BMVECs in a quiescent state and contributes to junction generation, and sphingosine-1 phosphate and activated protein C, which help balance junction proteins and cytoskeleton [182]. Immune cells also shift to anti-inflammatory phenotypes and develop mechanisms to diminish BBB breakdown. For example, anti-inflammatory astrocytes release pentraxin-3, which inhibits VEGF in the ischemic cerebral tissue and specifically supports BBB integrity [183]. Finally, neuronal progenitor cells migrate to the ischemic tissue to favor neurogenesis and neuroplasticity and restore BBB components [184,185].

4.4.2. Therapeutic Opportunities for MSCs Targeting the BBB in Brain Ischemia

Several studies have demonstrated the therapeutic effect of MSCs in preclinical models of stroke (Figure 5). Thus, understanding their effect on the BBB has become a topic of great interest in research [186].
For example, MSCs have shown the ability to protect neurons against oxidative damage. A study conducted by Huang et al. [187] showed that MSCs express high levels of antioxidant enzymes from the peroxiredoxin (PRDX) family. In this report, MSCs were able to rescue BBB integrity in an in vitro model of oxidative damage with the bEnd.3 cell line, by reducing TJ degradation and excessive ROS generation. This effect was found to be partially mediated by the secretion of PRDX4. Interestingly, silencing PRDX4 in MSCs attenuated their protective effect on BBB integrity, both in vitro and in the in vivo MCAO ischemic model. Conversely, Cheng et al. [188] investigated the effect of the treatment of MSCs administered 15 min after MCAO and found a reduction in IgG leakage in the brain parenchyma, along with the reversal of the TJ protein gap formation (ZO-1, occludin, and claudin-5). This treatment also suppressed MMP-9 upregulation, reduced neuroinflammation, and decreased neutrophil infiltration with downregulation of ICAM-1 expression. In a related study, Liu et al. [189] demonstrated that co-culture of MSCs with HUVECs in an in vitro ischemic-reperfusion model rescued injured endothelial cells via the generation of TNT-like structures, which was dependent on F-actin polymerization. They further investigated this effect in vivo in a model of MCAO in rats [190], in which the injection of MSCs 24 h after the ischemic induction led to a significant reduction in the infarct volume and higher microvessel densities in the peri-infarct areas. This was attributed to rescuing brain microvasculature through mitochondrial transfer with TNT formation in vivo. Moreover, Zacharek et al. [191] demonstrated that the administration of MSCs 24 h after MCAO in rats reduced BBB leakage and promoted angiogenesis and vascular stabilization in vivo and in vitro. This effect was achieved by increasing endogenous Ang-1/Tie2 and TJ proteins, and promoting the crosstalk between BMVECs and astrocytes.
Finally, different studies have demonstrated that the administration MSC-EVs can lead to a reduction in the infarct volume, improve neurological recovery, and enhanced angiogenesis. These effects are particularly significant when MSCs are exposed to hypoxic conditions prior to EV isolation [192]. These results suggest that it is not necessary to use the whole MSC for the treatment of ischemic stroke [193,194], evidencing MSC-EVs as a promising alternative for stroke therapy. Overall, these studies suggest that MSC treatment has a significant impact on BBB integrity and a great potential to modulate microvasculature and reduce the pathological processes associated with stroke.

5. Challenges and Future Directions

While preclinical studies have yielded promising results regarding the therapeutic potential of MSCs in neurodegenerative and neuroinflammatory disorders, the clinical efficacy of using MSCs remains uncertain and has produced mixed results (Table 2). A recent meta-analysis conducted by Kvistad et al. [195] aimed to assess the safety and efficacy of MSC therapy in various neuroinflammatory conditions, encompassing MS, ischemic stroke, and traumatic spinal cord injury. The findings indicated that the treatment was generally safe and well-tolerated by patients. However, the efficacy results were inconclusive, with no significant improvements. Irrespective of the neurodegenerative conditions discussed in this review, the limited efficacy observed in MSC treatment and the disparities between outcomes in mouse models and human clinical trials may be attributed to several factors. These include the specific source, culture, and isolation methods employed for MSCs, the dosage of transplanted cells, and the timing and route of delivery [195,196,197,198,199,200]. Furthermore, the accurate selection of patients and the identification of outcome measurements for assessing the success of MSC treatment are critical considerations that can significantly contribute to differences observed in clinical trials.
  • Source and culture of MSCs: MSCs from different sources such as BM-MSCs, ASCs, neural stem cells, and umbilical cells have been used in clinical trials (from both autologous and allogeneic origins), generating different results. These variations can be partially attributed to their distinct paracrine functions, leading to the secretion of different angiogenic, growth, and cytokine factors, which in turn influence their neuroprotective and immunological capabilities [201,202]. Furthermore, the methods used to harvest and isolate different MSCs can impact their yield, viability, and differentiation potential [203,204]. Depending on the source and intended use of the (allogeneic versus autologous) MSCs, the need of an extensive culturing process can also increase the senescence of MSCs, ultimately affecting the proliferative rates and their therapeutic efficacy [205,206]. Thus, significant differences exist in the methodological approaches used in the culture of MSCs, including culture reagents, cell expansion, cryopreservation, thawing procedures, fitness assessments, and functionality evaluations. As a result, there is a need for standardized protocols in the laboratory management of MSCs to mitigate inconsistencies across studies. In fact, the majority of human clinical trials employ allogeneic cryobanked MSCs, which are thawed immediately prior to transfusion. Cryopreservation methods and thawing protocols can also contribute to variations among clinical studies [207]. It is important to note that MSCs display molecular signs of cell injury in the first 24 h following retrieval from cryostorage. These molecular changes correlate with defects in suppression function in vitro, increased susceptibility to immune cell lysis, as well as reduced persistence in vivo following intravenous transfusion [208]. Allogeneic human MSCs typically transfused into patients within a few hours post thaw, directly retrieved from cryostorage, probably exhibit reduced viability, functionality, and in vivo persistence, compared to the cells routinely used in analogous murine systems. Additionally, patient-specific factors such as age, gender, genetic traits, existing co-morbidities, and the systemic effects of medications must be considered in autologous MSC treatments [197,209]. Interestingly, while autologous MSCs have been extensively used in most of the trials for AD, PD, and MS, their prolonged culture times needed for cell expansion make them less suitable for acute tissue injury conditions like stroke. In such conditions, allogeneic MSCs become the more feasible strategy [210]. Specifically, the clinical trials using allogeneic ASCs against neurodegenerative disorders have significantly increased in the last years (Table 2). These cells are abundant in adults and easy to isolate and expand. Furthermore, ASCs offer advantages for allogenic treatments, as they can be used to create a standardized and cost-effective donor bank, avoiding the issues associated with autologous treatments such as donor-recipient compatibility. However, it is important to note that during in vitro expansion major HLA class II molecules may increase in expression [211]. Allogeneic MSCs, when exposed to serum, can be vulnerable to complement-mediated injury, leading to reduced viability after infusion compared to autologous MSCs [212]. Therefore, in scenarios where long-term repeated administrations are necessary to impact outcomes for chronic disorders, the examination of immunological compatibility between donor MSCs and recipients may extend the survival and effectiveness of the MSCs [213].
  • MSC dose, timing and delivery: The challenges and disparities observed in clinical trials are also closely related to significant differences in cell doses and transplantation timing between laboratory settings and clinical practice. For instance, in preclinical studies involving stroke, an effective intravenous dose typically amounts to around four million cells for a rat weighing 250 g. This would translate to approximately 840 million cells in a stroke patient weighing 75 kg [214]. However, most clinical trials use doses considerably below this efficacious threshold, which may explain the observed lack of efficacy [215,216,217]. It is worth noting that stroke patients who received doses aligned with the findings of these preclinical studies displayed clinical improvements [218]. In addition, considerations regarding the administration of single or multiple doses, as well as the timing of these injections, can also result in significant differences in outcomes. Standardization of dosage is crucial to reduce variability between trials and gain insights into areas that require improvement. Additionally, the optimal route for stem cell injection can vary depending on the nature of the disease. For instance, in the case of MS, which is a multifocal and systemic disease, intravenous administration offers a straightforward and safe way to modulate aggressive immune responses in peripheral lymphatic tissues. A more localized distribution is desirable in conditions like PD, AD, and stroke after MSC delivery. However, in all of these disorders, despite the benefits observed when MSCs are intravenously injected in animal models, less successful therapeutic outcomes are often observed in clinical settings. This discrepancy is likely due to the entrapment of MSCs in systemic organs and the challenges associated with transmigration across the blood-brain barrier [219,220]. While intra-arterial administration of MSCs could serve as an alternative to intravenous administration, this approach increases the risk of embolic events [221,222]. The procoagulation status associated with MSCs and their size, which can lead to their arrest in small-diameter vessels, potentially causing vascular occlusion and reduced cerebral blood flow, may explain the risk of lethal pulmonary thromboembolism [223]. On the other hand, direct injection via intrathecal or intraventricular routes has shown promise [224,225]. While no significant differences were detected between intravenous and intrathecal routes in terms of MSC efficacy in patients with conditions like MS [226], some studies suggest that intrathecal administration of MSCs may be more efficacious than the intravenous route [227,228]. An alternative non-invasive and rapid delivery route for treating CNS disorders is intranasal administration of MSCs. Finally, preclinical models have demonstrated successful delivery of MSCs to the brain through this route, with therapeutic efficacy observed in conditions such as PD, MS, AD, and stroke [229,230,231,232].
  • Selection of patients: A significant source of variability in the outcomes of clinical trials stems from the heterogeneity of patient populations. This heterogeneity encompasses factors such as disease onset, development, stages (in AD, PD, and MS), and pathological phases of the disease (in stroke), disease severity, and the presence of co-morbidities. Therefore, careful consideration must be given to the rational selection of optimal candidate patients. For instance, in conditions like stroke, different phases of the pathological ischemic process offer distinct targets for MSC therapy, with varying beneficial effects. Early-phase trials have shown that MSC administration in stroke patients is safe and can reduce the inflammatory response, regulate the dynamic environment against toxicity, and decrease injury in the peri-infarct area [233,234]. However, the effectiveness of MSC therapy in clinically subacute and chronic ischemic stroke cases is yet to be fully validated. In MS, stem cell transplantation is more efficient when performed in the early stages of the disease because, in the later phases, after the emergence of chronic lesions, endogenous repair processes become compromised [235]. While endogenous repair mechanisms and injected MSCs can work in synergy during the repair of active plaques in relapsing-remitting MS, progressive forms of the disease witness a reduction in the function of reparative cells due to gliosis and the formation of fibrotic scars [236]. Similarly, in AD and PD, despite promising preclinical data, most clinical trials involving MSCs have yielded disappointing results due to the pathological variability corresponding to different stages of the diseases [196,198]. This heterogeneity in disease stages and phases introduces considerable variability across trials, both in terms of the therapeutic windows (spanning from acute to chronic stroke stages) and the routes of administration [215,216,217,237,238,239]. To address these challenges, the development of specific animal models with interventions tailored to particular disease stages will be essential for the effective clinical translation of novel therapeutics.
  • Clinical outcomes: Developing uniform and standardized measurements for assessing the efficacy of stem cell therapy is indeed crucial for reducing clinical failure. In diseases like PD and AD, where early-stage diagnostic tests are lacking, disease identification is often based on symptoms that manifest in the later stages, when a substantial portion of neurons has already been damaged, or through post-mortem pathological examinations. Consequently, the classification and staging of these diseases can vary significantly, leading to a high degree of heterogeneity in defined clinical groups. Indeed, achieving a more objective and standardized approach to patient selection and treatment evaluation is essential. This can be facilitated by the incorporation of various objective measures, including functional magnetic resonance imaging (fMRI), magnetic resonance tractography, and the use of blood and cerebral fluid biomarkers [240,241]. These measures not only may aid in identifying clinical improvements but also contribute to a more accurate assessment of disease progression and response to treatment. Furthermore, the timing of follow-up assessments is another critical consideration. Using more uniform clinical grouping criteria and outcome measures is pivotal for effectively evaluating the outcomes of cell therapy treatments. By adopting standardized assessment tools and criteria, researchers and clinicians can better compare results across different trials and refine treatment strategies for improved patient outcomes.
In summary, the valuable insights gained from clinical trials should guide future directions in stem cell therapy and underscore the critical need for a systematic approach to identify optimal conditions that can yield reliable and effective treatments. Several key strategies can contribute to achieving this goal, such as: (i) developing live imaging techniques to track and assess the bioavailability and biodistribution of MSCs in the body can provide valuable real-time information about their behavior and interactions; (ii) standardizing isolation and expansion protocols for MSCs that will help to reduce variability and ensure consistent quality in cell manufacturing; (iii) the incorporation of clear and reliable biomarkers that accurately reflect the therapeutic effects of stem cells in patients and thus can provide objective measures of treatment efficacy; and (iv) conducting well-designed randomized controlled studies that will help to establish the safety and efficacy of stem cell-based therapies while minimizing bias and confounding factors. To address the challenges and improve the clinical outcomes of MSC therapy in neuroinflammatory pathologies, it is imperative to gain a clearer understanding of the underlying biology and mechanisms of action of MSCs as well as to identify new cellular and/or molecular targets involved in these diseases. This knowledge will serve as a foundation for optimizing the treatment approach and developing targeted strategies that can enhance therapeutic efficacy. Regarding this, MSC priming or preconditioning approaches that expose cells to growth conditions mimicking the in vivo microenvironment of damaged tissue can improve the efficiency of MSC-mediated therapy. Various priming methods, such as using inflammatory cytokines, hypoxia, pharmacological drugs, chemical agents, biomaterials, and specific culture conditions, have shown promise [242]. However, it is important to note that the challenge lies in achieving consensus on cell manufacturing protocols for priming, which can ensure quality assurance for clinical-grade MSCs [243]. Additionally, as discussed in this review, the therapeutic potential of MSCs extends beyond their homing and differentiation capabilities. Their paracrine secretion of various factors, including trophic factors, immunomodulatory molecules, microvesicles, microRNAs, and mitochondrial transfer, plays a central role in their therapeutic effects. Recent investigations have explored cell-free therapies as promising approaches in neurodegeneration. Studies have shown that systemic administration of MSC-EVs can lead to functional improvement, offering a similar functional outcome with an improved safety profile compared to MSC administration, particularly in preclinical stroke models [244]. The use of allogeneic MSC-EVs confers several advantages, including low toxicity, low immunogenicity, enhanced safety, ease of injection, and improved ability to trespass biological barriers avoiding undesired entrapment in the lung microvasculature, and advantages in scalable production and storage [245]. However, despite promising preclinical results, only a few clinical studies on the effects of EV therapy in humans have been reported. These studies, while limited in scale, suggest that MSC-EVs are safe and may improve patient outcomes. Nevertheless, larger randomized trials are needed to thoroughly investigate the efficacy and safety of MSC-EVs therapy [246,247,248]. It is important to note that the cargo contents of MSC-EVs may exhibit age-dependent differences [249]. Several critical considerations must be addressed before the widespread clinical application of EVs. These include the establishment of specific guidelines for EV-based therapeutics, standardization of EV characterization, isolation, and storage methods, quality control requirements, and in-depth in vivo analyses of EV functionality. Variability in EV functional properties arising from different culture conditions is a crucial factor to consider for successful clinical translation.
Finally, recognizing that MSCs not only impact immune activation and neurodegeneration but also address disrupted vascular components has prompted the exploration of novel therapeutic targets and combination strategies. In addition to their impact on the BBB in the context of the CNS disorders covered in this review (PD, AD, MS, and stroke), the relevance of the BBB extends to other neurodegenerative conditions, including traumatic brain and spinal cord injury [250,251,252], amyotrophic lateral sclerosis [253,254], Huntington’s disease [255], and epilepsy [256]. However, the significance of the BBB as a target for MSC therapy in those disorders is still emerging. The understanding of the role of the BBB in the pathophysiology of neurodegenerative and neuroinflammatory diseases is pivotal for developing multi-target-based approaches. As discussed, the ability of MSCs to restore BBB integrity and functionality in aging-associated CNS pathologies such as AD, PD, MS, and stroke combined with their capacity to modulate the immune response and neurodegeneration, positions them as an attractive therapeutic option. This opens up the possibility of extending BBB targeting by MSC therapy to include other CNS-injured conditions. Prioritizing the BBB in the development of MSC-based therapies holds promise for enhancing clinical outcomes for patients suffering from these and other debilitating conditions.

Author Contributions

Conceptualization, E.G.-R., J.C.-G. and M.D.; writing—original draft preparation, E.G.-R., P.V.-R. and A.C.-M.; writing—review and editing, E.G.-R., J.C.-G., I.S.-M. and M.D.; supervision, E.G.-R. and R.M.L.; project administration, E.G.-R.; funding acquisition, E.G.-R. and R.M.L. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Spanish Ministry of Science and Innovation (PID2020-119638RB-I00 to E.G-R., PhD fellowship FPU17/02616 to J.C-G., and PRE2021-100172 to P.V-R) and Next Generation EU (Investigo—E-14-2023-0079369) to A.C-M.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

The following abbreviations are used in this manuscript:
AJsAdherens junctions
ASCsAdipose tissue-derived MSCs
ADAlzheimer’s disease
BMBasement membrane
BBBBlood-brain barrier
BM-MSCsBone marrow-derived mesenchymal stem cells
BMVECsBrain microvascular endothelial cells
CNSCentral nervous system
CCL2Chemokine (C-C motif) ligand 2
Extracellular amyloid-β
ICAM1Intercellular adhesion molecule 1
ILInterleukin
MSCMesenchymal Stem Cells
MSC-EVsMSC-derived extracellular vesicles
MCAOmiddle cerebral artery occlusion
MSMultiple sclerosis
NGFNerve growth factor
NVUNeurovascular unit
PDParkinson’s disease
PRDXPeroxiredoxin
PPMS/SPMSPrimary/secondary progressive MS
RRMSRelapsing-remitting MS
sRAGESecreted isoform of RAGE
SNpcSubstantia nigra pars compacta
TJsTight junctions
TIMPsTissue inhibitor metalloproteinases
VEGFVascular endothelial growth factor
WJWharton’s jelly

References

  1. WHO Reveals Reading Cause of Death and Disability Worldwide: 2000–2019. Available online: https://www.who.int/news/item/09-12-2020-who-reveals-leading-causes-of-death-and-disability-worldwide-2000-2019 (accessed on 6 May 2023).
  2. Banks, W.A.; Reed, M.J.; Logsdon, A.F.; Rhea, E.M.; Erickson, M.A. Healthy aging and the blood-brain barrier. Nat. Aging 2021, 1, 243–254. [Google Scholar] [CrossRef] [PubMed]
  3. Farrall, A.J.; Wardlaw, J.M. Blood-brain barrier: Ageing and microvascular disease--systematic review and meta-analysis. Neurobiol. Aging 2009, 30, 337–352. [Google Scholar] [CrossRef] [PubMed]
  4. Fiala, M.; Liu, Q.N.; Sayre, J.; Pop, V.; Brahmandam, V.; Graves, M.C.; Vinters, H.V. Cyclooxygenase-2-positive macrophages infiltrate the Alzheimer’s disease brain and damage the blood-brain barrier. Eur. J. Clin. Investig. 2002, 32, 360–371. [Google Scholar] [CrossRef] [PubMed]
  5. Elahy, M.; Jackaman, C.; Mamo, J.C.; Lam, V.; Dhaliwal, S.S.; Giles, C.; Nelson, D.; Takechi, R. Blood-brain barrier dysfunction developed during normal aging is associated with inflammation and loss of tight junctions but not with leukocyte recruitment. Immun. Ageing 2015, 12, 2. [Google Scholar] [CrossRef] [PubMed]
  6. Stamatovic, S.M.; Martinez, G.R.; Hu, A.; Choi, J.; Keep, R.F.; Andjelkovic, A.V. Decline in sirtuin-1 expression and activity plays a critical role in blood-brain barrier permeability in aging. Neurobiol. Dis. 2019, 126, 105–116. [Google Scholar] [CrossRef]
  7. Verheggen, I.C.M.; de Jong, J.J.A.; van Boxtel, M.P.J.; Postma, A.A.; Jansen, J.F.A.; Verhey, F.R.J.; Backes, W.H. Imaging the role of blood-brain barrier disruption in normal cognitive ageing. Geroscience 2020, 42, 1751–1764. [Google Scholar] [CrossRef] [PubMed]
  8. Verheggen, I.C.M.; de Jong, J.J.A.; van Boxtel, M.P.J.; Gronenschild, E.H.B.M.; Palm, W.M.; Postma, A.A.; Jansen, J.F.A.; Verhey, F.R.J.; Backes, W.H. Increase in blood-brain barrier leakage in healthy, older adults. Geroscience 2020, 42, 1183–1193. [Google Scholar] [CrossRef]
  9. Marchi, N.; Cavaglia, M.; Fazio, V.; Bhudia, S.; Hallene, K.; Janigro, D. Peripheral markers of blood-brain barrier damage. Clin. Chim. Acta 2004, 342, 1–12. [Google Scholar] [CrossRef]
  10. Mooradian, A.D.; Morin, A.M.; Cipp, L.J.; Haspel, H.C. Glucose transport is reduced in the blood-brain barrier of aged rats. Brain Res. 1991, 551, 145–149. [Google Scholar] [CrossRef]
  11. Banks, W.A.; Moinuddin, A.; Morley, J.E. Regional transport of TNF-alpha across the blood-brain barrier in young ICR and young and aged SAMP8 mice. Neurobiol. Aging 2001, 22, 671–676. [Google Scholar] [CrossRef]
  12. Staff, N.P.; Jones, D.T.; Singer, W. Mesenchymal stromal cell therapies for neurodegenerative diseases. Mayo Clin. Proc. 2019, 94, 892–905. [Google Scholar] [CrossRef] [PubMed]
  13. Andrzejewska, A.; Dabrowska, S.; Lukomska, B.; Janowski, M. Mesenchymal stem cells for neurological disorders. Adv. Sci. 2021, 8, 2002944. [Google Scholar] [CrossRef] [PubMed]
  14. Daneman, R.; Prat, A. The blood-brain barrier. Cold Spring Harb. Perspect. Biol. 2015, 7, a020412. [Google Scholar] [CrossRef] [PubMed]
  15. Persidsky, Y.; Ramirez, S.H.; Haorah, J.; Kanmogne, G.D. Blood-brain barrier: Structural components and function under physiologic and pathologic conditions. J. Neuroimmune Pharmacol. 2006, 1, 223–236. [Google Scholar] [CrossRef]
  16. Uwamori, H.; Ono, Y.; Yamashita, T.; Arai, K.; Sudo, R. Comparison of organ-specific endothelial cells in terms of microvascular formation and endothelial barrier functions. Microvasc. Res. 2019, 122, 60–70. [Google Scholar] [CrossRef]
  17. Hashimoto, Y.; Greene, C.; Munnich, A.; Campbell, M. The CLDN5 gene at the blood-brain barrier in health and disease. Fluids Barriers CNS 2023, 20, 1–22. [Google Scholar] [CrossRef]
  18. Wong, A.D.; Ye, M.; Levy, A.F.; Rothstein, J.D.; Bergles, D.E.; Searson, P.C. The blood-brain barrier: An engineering perspective. Front. Neuroeng. 2013, 6, 7. [Google Scholar] [CrossRef]
  19. Oldendorf, W.H.; Cornford, M.E.; Brown, W.J. The large apparent work capability of the blood-brain barrier: A study of the mitochondrial content of capillary endothelial cells in brain and other tissues of the rat. Ann. Neurol. 1977, 1, 409–417. [Google Scholar] [CrossRef]
  20. Paolinelli, R.; Corada, M.; Orsenigo, F.; Dejana, E. The Molecular Basis of the blood brain barrier differentiation and maintenance. Is it still a mystery? Pharmacol. Res. 2011, 63, 165–171. [Google Scholar] [CrossRef]
  21. Artus, C.; Glacial, F.; Ganeshamoorthy, K.; Ziegler, N.; Godet, M.; Guilbert, T.; Liebner, S.; Couraud, P.-O. The Wnt/planar cell polarity signaling pathway contributes to the integrity of tight junctions in brain endothelial cells. J. Cereb. Blood Flow. Metab. 2014, 34, 433–440. [Google Scholar] [CrossRef]
  22. Ghandour, M.S.; Langley, O.K.; Varga, V. Immunohistological localization of γ glutamyltranspeptidase in cerebellum at light and electron microscope levels. Neurosci. Lett. 1980, 20, 125–129. [Google Scholar] [CrossRef] [PubMed]
  23. Vorbrodt, A.W.; Lossinsky, A.S.; Wisniewski, H.M. Localization of alkaline phosphatase activity in endothelia of developing and mature mouse blood-brain barrier. Dev. Neurosci. 1986, 8, 1–13. [Google Scholar] [CrossRef] [PubMed]
  24. Betz, A.L. Sodium transport in capillaries isolated from rat brain. J. Neurochem. 1983, 41, 1150–1157. [Google Scholar] [CrossRef] [PubMed]
  25. van Tilborg, E.; van Kammen, C.M.; de Theije, C.G.M.; van Meer, M.P.A.; Dijkhuizen, R.M.; Nijboer, C.H. A quantitative method for microstructural analysis of myelinated axons in the injured rodent brain. Sci. Rep. 2017, 7, 16492. [Google Scholar] [CrossRef]
  26. Sweeney, M.D.; Zhao, Z.; Montagne, A.; Nelson, A.R.; Zlokovic, B.V. Blood-brain barrier: From physiology to disease and back. Physiol. Rev. 2019, 99, 21–78. [Google Scholar] [CrossRef]
  27. Badaut, J.; Bix, G.J. Vascular neural network phenotypic transformation after traumatic injury: Potential role in long-term sequelae. Transl. Stroke Res. 2014, 5, 394–406. [Google Scholar] [CrossRef]
  28. Sweeney, M.D.; Kisler, K.; Montagne, A.; Toga, A.W.; Zlokovic, B.V. The role of brain vasculature in neurodegenerative disorders. Nat. Neurosci. 2018, 21, 1318–1331. [Google Scholar] [CrossRef]
  29. Parker, K.R.; Migliorini, D.; Perkey, E.; Yost, K.E.; Bhaduri, A.; Bagga, P.; Haris, M.; Wilson, N.E.; Liu, F.; Gabunia, K.; et al. Single-cell analyses identify brain mural cells expressing CD19 as potential off-tumor targets for CAR-T immunotherapies. Cell 2020, 183, 126–142.e17. [Google Scholar] [CrossRef]
  30. Herndon, J.M.; Tome, M.E.; Davis, T.P. Primer on cerebrovascular diseases (Second Edition). In Primer on Cerebrovascular Diseases, 2nd ed.; p. Chapter 9-Development and Maintenance of the Blood-Brain Barrier; Academic Press: Cambridge, MA, USA, 2017; p. 53. ISBN 978-0-12-803059-2. [Google Scholar]
  31. Abbott, N.J.; Rönnbäck, L.; Hansson, E. Astrocyte-endothelial interactions at the blood-brain barrier. Nat. Rev. Neurosci. 2006, 7, 41–53. [Google Scholar] [CrossRef]
  32. Fallier-Becker, P.; Sperveslage, J.; Wolburg, H.; Noell, S. The Impact of agrin on the formation of orthogonal arrays of particles in cultured astrocytes from wild-type and agrin-null mice. Brain Res. 2011, 1367, 2–12. [Google Scholar] [CrossRef]
  33. Attwell, D.; Buchan, A.M.; Charpak, S.; Lauritzen, M.; Macvicar, B.A.; Newman, E.A. Glial and neuronal control of brain blood flow. Nature 2010, 468, 232–243. [Google Scholar] [CrossRef]
  34. Argaw, A.T.; Gurfein, B.T.; Zhang, Y.; Zameer, A.; John, G.R. VEGF-mediated disruption of endothelial CLN-5 promotes blood-brain barrier breakdown. Proc. Natl. Acad. Sci. USA 2009, 106, 1977–1982. [Google Scholar] [CrossRef]
  35. Hernández-Guillamon, M.; Delgado, P.; Ortega, L.; Pares, M.; Rosell, A.; García-Bonilla, L.; Fernández-Cadenas, I.; Borrell-Pagès, M.; Boada, M.; Montaner, J. Neuronal TIMP-1 release accompanies astrocytic MMP-9 secretion and enhances astrocyte proliferation induced by beta-amyloid 25–35 fragment. J. Neurosci. Res. 2009, 7, 2115–2125. [Google Scholar] [CrossRef]
  36. Owens, T.; Bechmann, I.; Engelhardt, B. Perivascular spaces and the two steps to neuroinflammation. J. Neuropathol. Exp. Neurol. 2008, 67, 1113–1121. [Google Scholar] [CrossRef] [PubMed]
  37. Wu, C.; Ivars, F.; Anderson, P.; Hallmann, R.; Vestweber, D.; Nilsson, P.; Robenek, H.; Tryggvason, K.; Song, J.; Korpos, E.; et al. Endothelial basement membrane laminin alpha5 selectively inhibits T lymphocyte extravasation into the brain. Nat. Med. 2009, 15, 519–527. [Google Scholar] [CrossRef] [PubMed]
  38. Korpos, E.; Wu, C.; Song, J.; Hallmann, R.; Sorokin, L. Role of the extracellular matrix in lymphocyte migration. Cell Tissue Res. 2010, 339, 47–57. [Google Scholar] [CrossRef]
  39. Xu, L.; Nirwane, A.; Yao, Y. Basement Membrane and blood–brain barrier. Stroke Vasc. Neurol. 2018, 4, 78–82. [Google Scholar] [CrossRef] [PubMed]
  40. Pardridge, W.M. Drug Transport across the blood-brain barrier. J. Cereb. Blood Flow. Metab. 2012, 32, 1959–1972. [Google Scholar] [CrossRef]
  41. Barar, J.; Rafi, M.A.; Pourseif, M.M.; Omidi, Y. Blood-brain barrier transport machineries and targeted therapy of brain diseases. Bioimpacts 2016, 6, 225–248. [Google Scholar] [CrossRef]
  42. Schulze, C.; Firth, J.A. Immunohistochemical localization of adherens junction components in blood-brain barrier microvessels of the rat. J. Cell Sci. 1993, 104 Pt 3, 773–782. [Google Scholar] [CrossRef]
  43. Dejana, E.; Tournier-Lasserve, E.; Weinstein, B.M. The Control of Vascular Integrity by Endothelial Cell Junctions: Molecular Basis and Pathological Implications. Dev. Cell 2009, 16, 209–221. [Google Scholar] [CrossRef] [PubMed]
  44. Tietz, S.; Engelhardt, B. Brain Barriers: Crosstalk between complex tight junctions and adherens junctions. J. Cell Biol. 2015, 209, 493–506. [Google Scholar] [CrossRef] [PubMed]
  45. Ueda, H.; Baba, T.; Terada, N.; Kato, Y.; Fujii, Y.; Takayama, I.; Mei, X.; Ohno, S. Immunolocalization of dystrobrevin in the astrocytic endfeet and endothelial cells in the rat cerebellum. Neurosci. Lett. 2000, 283, 121–124. [Google Scholar] [CrossRef] [PubMed]
  46. Dejana, E.; Vestweber, D. The Role of VE-Cadherin in vascular morphogenesis and permeability control. Prog. Mol. Biol. Transl. Sci. 2013, 116, 119–144. [Google Scholar] [CrossRef] [PubMed]
  47. Tzima, E.; Irani-Tehrani, M.; Kiosses, W.B.; Dejana, E.; Schultz, D.A.; Engelhardt, B.; Cao, G.; Delisser, H.; Schwartz, M.A. A mechanosensory complex that mediates the endothelial cell response to fluid shear stress. Nature 2005, 437, 426–431. [Google Scholar] [CrossRef]
  48. Turowski, P.; Martinelli, R.; Crawford, R.; Wateridge, D.; Papageorgiou, A.-P.; Lampugnani, M.G.; Gamp, A.C.; Vestweber, D.; Adamson, P.; Dejana, E.; et al. Phosphorylation of vascular endothelial cadherin controls lymphocyte emigration. J. Cell Sci. 2008, 121, 29–37. [Google Scholar] [CrossRef]
  49. Dickstein, D.L.; Biron, K.E.; Ujiie, M.; Pfeifer, C.G.; Jeffries, A.R.; Jefferies, W.A. Aβ peptide immunization restores blood-brain barrier integrity in Alzheimer disease. FASEB J. 2006, 20, 426–433. [Google Scholar] [CrossRef]
  50. Wessel, F.; Winderlich, M.; Holm, M.; Frye, M.; Rivera-Galdos, R.; Vockel, M.; Linnepe, R.; Ipe, U.; Stadtmann, A.; Zarbock, A.; et al. Leukocyte extravasation and vascular permeability are each controlled in vivo by different tyrosine residues of VE-Cadherin. Nat. Immunol. 2014, 15, 223–230. [Google Scholar] [CrossRef]
  51. Takeichi, M. Dynamic contacts: Rearranging adherens junctions to drive epithelial remodelling. Nat. Rev. Mol. Cell Biol. 2014, 15, 397–410. [Google Scholar] [CrossRef]
  52. Nicholson, B.J.; Weber, P.A.; Cao, F.; Chang, H.C.; Lampe, P.; Goldberg, G. The molecular basis of selective permeability of connexins is complex and includes both size and charge. Braz. J. Med. Biol. Res. 2000, 33, 369–398. [Google Scholar] [CrossRef]
  53. Stamatovic, S.M.; Johnson, A.M.; Keep, R.F.; Andjelkovic, A.V. Junctional proteins of the blood-brain barrier: New insight into function and dysfunction. Tissue Barriers 2016, 4, e1154641. [Google Scholar] [CrossRef] [PubMed]
  54. Ezan, P.; André, P.; Cisternino, S.; Saubaméa, B.; Boulay, A.-C.; Doutremer, S.; Thomas, M.-A.; Quenech’du, N.; Giaume, C.; Cohen-Salmon, M. Deletion of astroglial connexins weakens the blood–brain barrier. J. Cereb. Blood Flow. Metab. 2012, 32, 1457. [Google Scholar] [CrossRef]
  55. Kotini, M.; Barriga, E.H.; Leslie, J.; Gentzel, M.; Rauschenberger, V.; Schambony, A.; Mayor, R. Gap junction protein Connexin-43 is a direct transcriptional regulator of N-cadherin in vivo. Nat. Commun. 2018, 9, 3846. [Google Scholar] [CrossRef] [PubMed]
  56. Devraj, K.; Klinger, M.E.; Myers, R.L.; Mokashi, A.; Hawkins, R.A.; Simpson, I.A. GLUT-1 Glucose transporters in the blood-brain barrier: Differential phosphorylation. J. Neurosci. Res. 2011, 89, 1913–1925. [Google Scholar] [CrossRef] [PubMed]
  57. Pines, G.; Danbolt, N.C.; Bjørås, M.; Zhang, Y.; Bendahan, A.; Eide, L.; Koepsell, H.; Storm-Mathisen, J.; Seeberg, E.; Kanner, B.I. Cloning and expression of a rat brain L-Glutamate transporter. Nature 1992, 360, 464–467. [Google Scholar] [CrossRef]
  58. Huttunen, J.; Peltokangas, S.; Gynther, M.; Natunen, T.; Hiltunen, M.; Auriola, S.; Ruponen, M.; Vellonen, K.S.; Huttunen, K.M. L-Type Amino Acid Transporter 1 (LAT1/Lat1)-Utilizing prodrugs can improve the delivery of drugs into neurons, astrocytes and microglia. Sci. Rep. 2019, 9, 12860. [Google Scholar] [CrossRef]
  59. Mann, G.E.; Yudilevich, D.L.; Sobrevia, L. Regulation of amino acid and glucose transporters in endothelial and smooth muscle cells. Physiol. Rev. 2003, 83, 183–252. [Google Scholar] [CrossRef]
  60. O’Kane, R.L.; Martínez-López, I.; DeJoseph, M.R.; Viña, J.R.; Hawkins, R.A. Na+-Dependent glutamate transporters (EAAT1, EAAT2, and EAAT3) of the blood-brain barrier: A mechanism for glutamate removal. J. Biol. Chem. 1999, 274, 31891–31895. [Google Scholar] [CrossRef]
  61. Simpson, I.A.; Carruthers, A.; Vannucci, S.J. Supply and demand in cerebral energy metabolism: The role of nutrient transporters. J. Cereb. Blood Flow. Metab. 2007, 27, 1766–1791. [Google Scholar] [CrossRef]
  62. Vijay, N.; Morris, M.E. Role of monocarboxylate transporters in drug delivery to the brain. Curr. Pharm. Des. 2014, 20, 1487–1498. [Google Scholar] [CrossRef]
  63. Leclerc, M.; Bourassa, P.; Tremblay, C.; Caron, V.; Sugère, C.; Emond, V.; Bennett, D.A.; Calon, F. Cerebrovascular insulin receptors are defective in Alzheimer’s Disease. Brain 2023, 146, 75–90. [Google Scholar] [CrossRef]
  64. Lillis, A.P.; Van Duyn, L.B.; Murphy-Ullrich, J.E.; Strickland, D.K. LDL Receptor-related protein 1: Unique tissue-specific functions revealed by selective gene knockout studies. Physiol. Rev. 2008, 88, 887–918. [Google Scholar] [CrossRef] [PubMed]
  65. Deane, R.; Du Yan, S.; Submamaryan, R.K.; LaRue, B.; Jovanovic, S.; Hogg, E.; Welch, D.; Manness, L.; Lin, C.; Yu, J.; et al. RAGE mediates Amyloid-beta peptide transport across the blood-brain barrier and accumulation in brain. Nat. Med. 2003, 9, 907–913. [Google Scholar] [CrossRef] [PubMed]
  66. Giridharan, V.V.; Generoso, J.S.; Collodel, A.; Dominguini, D.; Faller, C.J.; Tardin, F.; Bhatti, G.S.; Petronilho, F.; Dal-Pizzol, F.; Barichello, T. Receptor for advanced glycation end products (RAGE) mediates cognitive impairment triggered by Pneumococcal Meningitis. Neurotherapeutics 2021, 18, 640–653. [Google Scholar] [CrossRef] [PubMed]
  67. Ahmed Juvale, I.I.; Abdul Hamid, A.A.; Abd Halim, K.B.; Che Has, A.T. P-Glycoprotein: New insights into structure, physiological function, regulation and alterations in disease. Heliyon 2022, 8, e09777. [Google Scholar] [CrossRef]
  68. Wei, W.; Bodles-Brakhop, A.M.; Barger, S.W. A Role for P-Glycoprotein in clearance of Alzheimer Amyloid β-Peptide from the brain. Curr. Alzheimer Res. 2016, 13, 615–620. [Google Scholar] [CrossRef]
  69. Bourassa, P.; Alata, W.; Tremblay, C.; Paris-Robidas, S.; Calon, F. Transferrin receptor-mediated uptake at the blood-brain barrier is not impaired by Alzheimer’s Disease neuropathology. Mol. Pharm. 2019, 16, 583–594. [Google Scholar] [CrossRef]
  70. Gimble, J.M.; Guilak, F.; Nuttall, M.E.; Sathishkumar, S.; Vidal, M.; Bunnell, B.A. In vitro differentiation potential of mesenchymal stem cells. Transfus. Med. Hemother. 2008, 35, 228–238. [Google Scholar] [CrossRef]
  71. Ryan, J.M.; Barry, F.P.; Murphy, J.M.; Mahon, B.P. Mesenchymal stem cells avoid allogeneic rejection. J. Inflamm. 2005, 2, 8. [Google Scholar] [CrossRef]
  72. Bourin, P.; Bunnell, B.A.; Casteilla, L.; Dominici, M.; Katz, A.J.; March, K.L.; Redl, H.; Rubin, J.P.; Yoshimura, K.; Gimble, J.M. Stromal cells from the adipose tissue-derived stromal vascular fraction and culture expanded adipose tissue-derived stromal/stem cells: A joint statement of the International Federation for Adipose Therapeutics and Science (IFATS) and the International Society for Cellular Therapy (ISCT). Cytotherapy 2013, 15, 641–648. [Google Scholar] [CrossRef]
  73. Crisan, M.; Yap, S.; Casteilla, L.; Chen, C.-W.; Corselli, M.; Park, T.S.; Andriolo, G.; Sun, B.; Zheng, B.; Zhang, L.; et al. A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell 2008, 3, 301–313. [Google Scholar] [CrossRef] [PubMed]
  74. Friedenstein, A.J.; Chailakhyan, R.K.; Gerasimov, U.V. Bone marrow osteogenic stem cells: In vitro cultivation and transplantation in diffusion chambers. Cell Tissue Kinet. 1987, 20, 263–272. [Google Scholar] [CrossRef] [PubMed]
  75. Hass, R.; Kasper, C.; Böhm, S.; Jacobs, R. Different populations and sources of human mesenchymal stem cells (MSC): A comparison of adult and neonatal tissue-derived MSC. Cell Commun. Signal 2011, 9, 12. [Google Scholar] [CrossRef] [PubMed]
  76. Fraser, J.K.; Wulur, I.; Alfonso, Z.; Hedrick, M.H. Fat Tissue: An underappreciated source of stem cells for biotechnology. Trends Biotechnol. 2006, 24, 150–154. [Google Scholar] [CrossRef]
  77. Liau, L.L.; Ruszymah, B.H.I.; Ng, M.H.; Law, J.X. Characteristics and clinical applications of Wharton’s Jelly-derived mesenchymal stromal cells. Curr. Res. Transl. Med. 2020, 68, 5–16. [Google Scholar] [CrossRef]
  78. ClinicalTrials.Gov. Available online: https://classic.clinicaltrials.gov (accessed on 26 July 2023).
  79. Cova, L.; Armentero, M.-T.; Zennaro, E.; Calzarossa, C.; Bossolasco, P.; Busca, G.; Lambertenghi Deliliers, G.; Polli, E.; Nappi, G.; Silani, V.; et al. Multiple neurogenic and neurorescue effects of human mesenchymal stem cell after transplantation in an experimental model of Parkinson’s Disease. Brain Res. 2010, 1311, 12–27. [Google Scholar] [CrossRef]
  80. Li, W.; Ma, N.; Ong, L.-L.; Nesselmann, C.; Klopsch, C.; Ladilov, Y.; Furlani, D.; Piechaczek, C.; Moebius, J.M.; Lützow, K.; et al. Bcl-2 engineered MSCs inhibited apoptosis and improved heart function. Stem Cells 2007, 25, 2118–2127. [Google Scholar] [CrossRef]
  81. Oh, S.H.; Kim, H.N.; Park, H.-J.; Shin, J.Y.; Lee, P.H. Mesenchymal stem cells increase hippocampal neurogenesis and neuronal differentiation by enhancing the Wnt signaling pathway in an Alzheimer’s Disease model. Cell Transplant. 2015, 24, 1097–1109. [Google Scholar] [CrossRef]
  82. Zhang, Y.; Yu, S.; Tuazon, J.P.; Lee, J.-Y.; Corey, S.; Kvederis, L.; Kingsbury, C.; Kaneko, Y.; Borlongan, C.V. Neuroprotective effects of human bone marrow mesenchymal stem cells against cerebral ischemia are mediated in part by an anti-apoptotic mechanism. Neural Regen. Res. 2019, 14, 597. [Google Scholar] [CrossRef]
  83. Paliwal, S.; Chaudhuri, R.; Agrawal, A.; Mohanty, S. Regenerative abilities of mesenchymal stem cells through mitochondrial transfer. J. Biomed. Sci. 2018, 25, 31. [Google Scholar] [CrossRef]
  84. Norat, P.; Soldozy, S.; Sokolowski, J.D.; Gorick, C.M.; Kumar, J.S.; Chae, Y.; Yağmurlu, K.; Prada, F.; Walker, M.; Levitt, M.R.; et al. Mitochondrial dysfunction in neurological disorders: Exploring mitochondrial transplantation. NPJ Regen. 2020, 5, 22. [Google Scholar] [CrossRef]
  85. Boukelmoune, N.; Chiu, G.S.; Kavelaars, A.; Heijnen, C.J. Mitochondrial transfer from mesenchymal stem cells to neural stem cells protects against the neurotoxic effects of cisplatin. Acta Neuropathol. Commun. 2018, 6, 139. [Google Scholar] [CrossRef] [PubMed]
  86. Wang, Q.; Yang, Q.; Wang, Z.; Tong, H.; Ma, L.; Zhang, Y.; Shan, F.; Meng, Y.; Yuan, Z. Comparative analysis of human mesenchymal stem cells from fetal-bone marrow, adipose tissue, and Warton’s Jelly as sources of cell immunomodulatory therapy. Hum. Vaccines Immunother. 2016, 12, 85–96. [Google Scholar] [CrossRef] [PubMed]
  87. Di Nicola, M.; Carlo-Stella, C.; Magni, M.; Milanesi, M.; Longoni, P.D.; Matteucci, P.; Grisanti, S.; Gianni, A.M. Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. Blood 2002, 99, 3838–3843. [Google Scholar] [CrossRef] [PubMed]
  88. François, M.; Romieu-Mourez, R.; Li, M.; Galipeau, J. Human MSC Suppression correlates with cytokine induction of indoleamine 2,3-Dioxygenase and bystander M2 macrophage differentiation. Mol. Ther. 2012, 20, 187–195. [Google Scholar] [CrossRef] [PubMed]
  89. Li, N.; Hua, J. Interactions between mesenchymal stem cells and the immune system. Cell. Mol. Life Sci. 2017, 74, 2345–2360. [Google Scholar] [CrossRef]
  90. Sato, K.; Ozaki, K.; Oh, I.; Meguro, A.; Hatanaka, K.; Nagai, T.; Muroi, K.; Ozawa, K. Nitric oxide plays a critical role in suppression of T-cell proliferation by mesenchymal stem cells. Blood 2007, 109, 228–234. [Google Scholar] [CrossRef]
  91. Liu, Y.; Zeng, R.; Wang, Y.; Huang, W.; Hu, B.; Zhu, G.; Zhang, R.; Li, F.; Han, J.; Li, Y. Mesenchymal stem cells enhance microglia M2 polarization and attenuate neuroinflammation through TSG-6. Brain Res. 2019, 1724, 146422. [Google Scholar] [CrossRef]
  92. Gu, Y.; He, M.; Zhou, X.; Liu, J.; Hou, N.; Bin, T.; Zhang, Y.; Li, T.; Chen, J. Endogenous IL-6 of mesenchymal stem cell improves behavioral outcome of hypoxic-ischemic brain damage neonatal rats by suppressing apoptosis in astrocyte. Sci. Rep. 2016, 6, 18587. [Google Scholar] [CrossRef]
  93. Kim, J.-Y.; Kim, D.H.; Kim, J.H.; Lee, D.; Jeon, H.B.; Kwon, S.-J.; Kim, S.M.; Yoo, Y.J.; Lee, E.H.; Choi, S.J.; et al. Soluble intracellular adhesion molecule-1 secreted by human umbilical cord blood-derived mesenchymal stem cell reduces Amyloid-β plaques. Cell Death Differ. 2012, 19, 680–691. [Google Scholar] [CrossRef]
  94. Sarkar, S. Protein aggregation in neurodegenerative disorders: A cause or consequence? Adv. Tech. Biol. Med. 2015, 2, e105. [Google Scholar] [CrossRef]
  95. Fischer, U.M.; Harting, M.T.; Jimenez, F.; Monzon-Posadas, W.O.; Xue, H.; Savitz, S.I.; Laine, G.A.; Cox, C.S. Pulmonary passage is a major obstacle for intravenous stem cell delivery: The pulmonary first-pass effect. Stem Cells Dev. 2009, 18, 683–692. [Google Scholar] [CrossRef] [PubMed]
  96. Rüster, B.; Göttig, S.; Ludwig, R.J.; Bistrian, R.; Müller, S.; Seifried, E.; Gille, J.; Henschler, R. Mesenchymal stem cells display coordinated rolling and adhesion behavior on endothelial cells. Blood 2006, 108, 3938–3944. [Google Scholar] [CrossRef] [PubMed]
  97. Yilmaz, G.; Vital, S.; Yilmaz, C.E.; Stokes, K.Y.; Alexander, J.S.; Granger, D.N. Selectin-mediated recruitment of bone marrow stromal cells in the postischemic cerebral microvasculature. Stroke 2011, 42, 806–811. [Google Scholar] [CrossRef] [PubMed]
  98. Mitkari, B.; Kerkelä, E.; Nystedt, J.; Korhonen, M.; Mikkonen, V.; Huhtala, T.; Jolkkonen, J. Intra-arterial infusion of human bone marrow-derived mesenchymal stem cells results in transient localization in the brain after cerebral ischemia in rats. Exp. Neurol. 2013, 239, 158–162. [Google Scholar] [CrossRef] [PubMed]
  99. Lee, N.K.; Yang, J.; Chang, E.H.; Park, S.E.; Lee, J.; Choi, S.J.; Oh, W.; Chang, J.W.; Na, D.L. Intra-arterially delivered mesenchymal stem cells are not detected in the brain parenchyma in an Alzheimer’s Disease mouse model. PLoS ONE 2016, 11, e0155912. [Google Scholar] [CrossRef]
  100. Albert, M.S. Changes in cognition. Neurobiol. Aging 2011, 32 (Suppl. S1), S58–S63. [Google Scholar] [CrossRef]
  101. Whitaker, K.W.; LaFerla, F.M.; Steinbusch, H.W.M.; Lemere, C.A.; Bovenkamp, D.E. BrightFocus Alzheimer’s fast track 2019. Mol. Neurodegener. 2019, 14, 48. [Google Scholar] [CrossRef]
  102. Terry, R.D. Neuropathological changes in Alzheimer disease. Prog. Brain Res. 1994, 101, 383–390. [Google Scholar] [CrossRef]
  103. Bertram, L.; Tanzi, R.E. Thirty years of Alzheimer’s disease genetics: The implications of systematic meta-analyses. Nat. Rev. Neurosci. 2008, 9, 768–778. [Google Scholar] [CrossRef]
  104. Carrano, A.; Hoozemans, J.J.M.; van der Vies, S.M.; van Horssen, J.; de Vries, H.E.; Rozemuller, A.J.M. Neuroinflammation and blood-brain barrier changes in capillary amyloid angiopathy. Neurodegener. Dis. 2012, 10, 329–331. [Google Scholar] [CrossRef] [PubMed]
  105. Marco, S.; Skaper, S.D. Amyloid Beta-peptide1-42 alters tight junction protein distribution and expression in brain microvessel endothelial cells. Neurosci. Lett. 2006, 401, 219–224. [Google Scholar] [CrossRef] [PubMed]
  106. Mooradian, A.D.; Chung, H.C.; Shah, G.N. GLUT-1 expression in the cerebra of patients with Alzheimer’s disease. Neurobiol. Aging 1997, 18, 469–474. [Google Scholar] [CrossRef] [PubMed]
  107. Hooijmans, C.R.; Graven, C.; Dederen, P.J.; Tanila, H.; van Groen, T.; Kiliaan, A.J. Amyloid beta deposition is related to decreased glucose transporter-1 levels and hippocampal atrophy in brains of aged APP/PS1 mice. Brain Res. 2007, 1181, 93–103. [Google Scholar] [CrossRef]
  108. Montagne, A.; Barnes, S.R.; Sweeney, M.D.; Halliday, M.R.; Sagare, A.P.; Zhao, Z.; Toga, A.W.; Jacobs, R.E.; Liu, C.Y.; Amezcua, L.; et al. Blood-brain barrier breakdown in the aging human hippocampus. Neuron 2015, 85, 296–302. [Google Scholar] [CrossRef]
  109. Wilcock, D.M.; Vitek, M.P.; Colton, C.A. Vascular amyloid alters astrocytic water and potassium channels in mouse models and humans with Alzheimer’s disease. Neuroscience 2009, 159, 1055–1069. [Google Scholar] [CrossRef]
  110. de Jong, G.I.; Jansen, A.S.; Horvath, E.; Gispen, W.H.; Luiten, P.G. Nimodipine effects on cerebral microvessels and sciatic nerve in aging rats. Neurobiol. Aging 1992, 13, 73–81. [Google Scholar] [CrossRef]
  111. Kalaria, R.N.; Pax, A.B. Increased collagen content of cerebral microvessels in Alzheimer’s disease. Brain Res. 1995, 705, 349–352. [Google Scholar] [CrossRef]
  112. Christov, A.; Ottman, J.; Hamdheydari, L.; Grammas, P. Structural changes in Alzheimer’s disease brain microvessels. Curr. Alzheimer Res. 2008, 5, 392–395. [Google Scholar] [CrossRef]
  113. Jung, S.S.; Zhang, W.; Van Nostrand, W.E. Pathogenic a beta induces the expression and activation of matrix metalloproteinase-2 in human cerebrovascular smooth muscle cells. J. Neurochem. 2003, 85, 1208–1215. [Google Scholar] [CrossRef]
  114. Asahina, M.; Yoshiyama, Y.; Hattori, T. Expression of matrix metalloproteinase-9 and urinary-type plasminogen activator in Alzheimer’s disease brain. Clin. Neuropathol. 2001, 20, 60–63. [Google Scholar] [PubMed]
  115. Miller, M.C.; Tavares, R.; Johanson, C.E.; Hovanesian, V.; Donahue, J.E.; Gonzalez, L.; Silverberg, G.D.; Stopa, E.G. Hippocampal RAGE immunoreactivity in early and advanced Alzheimer’s disease. Brain Res. 2008, 1230, 273–280. [Google Scholar] [CrossRef] [PubMed]
  116. van Assema, D.M.E.; Lubberink, M.; Bauer, M.; van der Flier, W.M.; Schuit, R.C.; Windhorst, A.D.; Comans, E.F.I.; Hoetjes, N.J.; Tolboom, N.; Langer, O.; et al. Blood-brain barrier p-glycoprotein function in Alzheimer’s disease. Brain 2012, 135, 181–189. [Google Scholar] [CrossRef] [PubMed]
  117. Shibata, M.; Yamada, S.; Kumar, S.R.; Calero, M.; Bading, J.; Frangione, B.; Holtzman, D.M.; Miller, C.A.; Strickland, D.K.; Ghiso, J.; et al. Clearance of Alzheimer’s amyloid-Ss(1-40) peptide from brain by LDL receptor-related protein-1 at the blood-brain barrier. J. Clin. Invest. 2000, 106, 1489–1499. [Google Scholar] [CrossRef] [PubMed]
  118. Deane, R.; Wu, Z.; Sagare, A.; Davis, J.; Du Yan, S.; Hamm, K.; Xu, F.; Parisi, M.; LaRue, B.; Hu, H.W.; et al. LRP/Amyloid beta-peptide interaction mediates differential brain efflux of Aβ isoforms. Neuron 2004, 43, 333–344. [Google Scholar] [CrossRef]
  119. Thomsen, M.S.; Routhe, L.J.; Moos, T. The vascular basement membrane in the healthy and pathological brain. J. Cereb. Blood Flow. Metab. 2017, 37, 3300–3317. [Google Scholar] [CrossRef]
  120. Hohsfield, L.A.; Humpel, C. Migration of blood cells to β-amyloid plaques in Alzheimer’s disease. Exp. Gerontol. 2015, 65, 8–15. [Google Scholar] [CrossRef]
  121. Zenaro, E.; Pietronigro, E.; Della Bianca, V.; Piacentino, G.; Marongiu, L.; Budui, S.; Turano, E.; Rossi, B.; Angiari, S.; Dusi, S.; et al. Neutrophils promote Alzheimer’s disease-like pathology and cognitive decline via LFA-1 integrin. Nat. Med. 2015, 21, 880–886. [Google Scholar] [CrossRef]
  122. Zhang, L.; Dong, Z.; Zhang, J. Immunomodulatory role of mesenchymal stem cells in Alzheimer’s disease. Life Sci. 2020, 246, 117405. [Google Scholar] [CrossRef]
  123. Garcia, K.; Ornellas, F.; Matsumoto, P.; Patti, C.; Mello, L.; Frussa-Filho, R.; Han, S.; Longo, B. Therapeutic effects of the transplantation of VEGF overexpressing bone marrow mesenchymal stem cells in the hippocampus of murine model of Alzheimer’s disease. Front. Aging Neurosci. 2014, 6, 30. [Google Scholar] [CrossRef]
  124. Son, M.; Oh, S.; Park, H.; Ahn, H.; Choi, J.; Kim, H.; Lee, H.S.; Lee, S.; Park, H.-J.; Kim, S.U.; et al. Protection against RAGE-mediated neuronal cell death by SRAGE-secreting human mesenchymal stem cells in 5xFAD transgenic mouse model. Brain Behav. Immun. 2017, 66, 347–358. [Google Scholar] [CrossRef] [PubMed]
  125. Liu, X.L.; Ouyang, F.B.; Hu, L.T.; Sun, P.; Yang, J.; Sun, Y.J.; Liao, M.S.; Lan, L.F.; Pei, Z.; Fan, Y.H. Mesenchymal stem cells improve cognitive impairment and reduce Aβ deposition via promoting AQP4 polarity and relieving neuroinflammation in rats with chronic hypertension-induced cerebral small-vessel disease. Front. Aging Neurosci. 2022, 14, 883503. [Google Scholar] [CrossRef] [PubMed]
  126. Tachibana, M.; Yamazaki, Y.; Liu, C.-C.; Bu, G.; Kanekiyo, T. Pericyte implantation in the brain enhances cerebral blood flow and reduces amyloid-β pathology in amyloid model mice. Exp. Neurol. 2018, 300, 13–21. [Google Scholar] [CrossRef] [PubMed]
  127. Kim, S.; Lee, S.; Lim, J.; Choi, H.; Kang, H.; Jeon, N.L.; Son, Y. Human bone marrow-derived mesenchymal stem cells play a role as a vascular pericyte in the reconstruction of human BBB on the angiogenesis microfluidic chip. Biomaterials 2021, 279, 121210. [Google Scholar] [CrossRef] [PubMed]
  128. Kang, S.G.; Shinojima, N.; Hossain, A.; Gumin, J.; Yong, R.L.; Colman, H.; Marini, F.; Andreeff, M.; Lang, F.F. Isolation and perivascular localization of mesenchymal stem cells from mouse brain. Neurosurgery 2010, 67, 711. [Google Scholar] [CrossRef] [PubMed]
  129. Poewe, W.; Seppi, K.; Tanner, C.M.; Halliday, G.M.; Brundin, P.; Volkmann, J.; Schrag, A.-E.; Lang, A.E. Parkinson disease. Nat. Rev. Dis. Primers 2017, 3, 17013. [Google Scholar] [CrossRef]
  130. Al-Bachari, S.; Naish, J.H.; Parker, G.J.M.; Emsley, H.C.A.; Parkes, L.M. Blood–brain barrier leakage is increased in Parkinson’s disease. Front. Physiol. 2020, 11, 593026. [Google Scholar] [CrossRef]
  131. Gray, M.T.; Woulfe, J.M. Striatal blood-brain barrier permeability in Parkinson’s disease. J. Cereb. Blood Flow. Metab. 2015, 35, 747–750. [Google Scholar] [CrossRef]
  132. Pienaar, I.S.; Lee, C.H.; Elson, J.L.; McGuinness, L.; Gentleman, S.M.; Kalaria, R.N.; Dexter, D.T. Deep-brain stimulation associates with improved microvascular integrity in the subthalamic nucleus in Parkinson’s disease. Neurobiol. Dis. 2015, 74, 392–405. [Google Scholar] [CrossRef]
  133. Loeffler, D.A.; Connor, J.R.; Juneau, P.L.; Snyder, B.S.; Kanaley, L.; DeMaggio, A.J.; Nguyen, H.; Brickman, C.M.; LeWitt, P.A. Transferrin and iron in normal, Alzheimer’s disease, and Parkinson’s disease brain regions. J. Neurochem. 1995, 65, 710–724. [Google Scholar] [CrossRef]
  134. Li, J.Q.; Tan, L.; Yu, J.T. The role of the LRRK2 gene in parkinsonism. Mol. Neurodegener. 2014, 9, 47. [Google Scholar] [CrossRef] [PubMed]
  135. Droździk, M.; Białecka, M.; Myśliwiec, K.; Honczarenko, K.; Stankiewicz, J.; Sych, Z. Polymorphism in the p-glycoprotein drug transporter MDR1 gene: A possible link between environmental and genetic factors in Parkinson’s disease. Pharmacogenetics 2003, 13, 259–263. [Google Scholar] [CrossRef] [PubMed]
  136. Faucheux, B.A.; Bonnet, A.M.; Agid, Y.; Hirsch, E.C. Blood vessels change in the mesencephalon of patients with Parkinson’s disease. Lancet 1999, 353, 981–982. [Google Scholar] [CrossRef] [PubMed]
  137. Barcia, C.; Emborg, M.E.; Hirsch, E.C.; Herrero, M.-T. Blood vessels and parkinsonism. Front. Biosci. 2004, 9, 277–282. [Google Scholar] [CrossRef]
  138. Desai, B.S.; Monahan, A.J.; Carvey, P.M.; Hendey, B. Blood-brain barrier pathology in Alzheimer’s and Parkinson’s disease: Implications for drug therapy. Cell Transpl. 2007, 16, 285–299. [Google Scholar] [CrossRef]
  139. Streit, W.J.; Mrak, R.E.; Griffin, W.S.T. Microglia and neuroinflammation: A pathological perspective. J. Neuroinflamm. 2004, 1, 14. [Google Scholar] [CrossRef]
  140. Wong, D.; Dorovini-Zis, K.; Vincent, S.R. Cytokines, nitric oxide, and CGMP modulate the permeability of an in vitro model of the human blood-brain barrier. Exp. Neurol. 2004, 190, 446–455. [Google Scholar] [CrossRef]
  141. Chao, Y.X.; He, B.P.; Tay, S.S.W. Mesenchymal stem cell transplantation attenuates blood brain barrier damage and neuroinflammation and protects dopaminergic neurons against MPTP toxicity in the substantia nigra in a model of Parkinson’s disease. J. Neuroimmunol. 2009, 216, 39–50. [Google Scholar] [CrossRef]
  142. Xue, C.; Li, X.; Ba, L.; Zhang, M.; Yang, Y.; Gao, Y.; Sun, Z.; Han, Q.; Zhao, R.C. MSC-derived exosomes can enhance the angiogenesis of human brain MECs and show therapeutic potential in a mouse model of Parkinson’s disease. Aging Dis. 2021, 12, 1211–1222. [Google Scholar] [CrossRef]
  143. Park, H.J.; Shin, J.Y.; Kim, H.N.; Oh, S.H.; Song, S.K.; Lee, P.H. Mesenchymal stem cells stabilize the blood–brain barrier through regulation of astrocytes. Stem Cell Res. Ther. 2015, 6, 187. [Google Scholar] [CrossRef]
  144. Reich, D.S.; Lucchinetti, C.F.; Calabresi, P.A. Multiple Sclerosis. N. Engl. J. Med. 2018, 378, 169–180. [Google Scholar] [CrossRef] [PubMed]
  145. Lassmann, H. Pathology and disease mechanisms in different stages of multiple sclerosis. J. Neurol. Sci. 2013, 333, 1–4. [Google Scholar] [CrossRef] [PubMed]
  146. Liddelow, S.A.; Guttenplan, K.A.; Clarke, L.E.; Bennett, F.C.; Bohlen, C.J.; Schirmer, L.; Bennett, M.L.; Münch, A.E.; Chung, W.-S.; Peterson, T.C.; et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 2017, 541, 481–487. [Google Scholar] [CrossRef]
  147. Schreiner, T.G.; Romanescu, C.; Popescu, B.O. The blood–brain barrier—A key player in multiple sclerosis disease mechanisms. Biomolecules 2022, 12, 538. [Google Scholar] [CrossRef] [PubMed]
  148. Yang, F.; Zhao, K.; Zhang, X.; Zhang, J.; Xu, B. ATP induces disruption of tight junction proteins via IL-1 beta-dependent MMP-9 activation of human blood-brain barrier in vitro. Neural Plast. 2016, 2016, 8928530. [Google Scholar] [CrossRef] [PubMed]
  149. Desai, T.R.; Leeper, N.J.; Hynes, K.L.; Gewertz, B.L. Interleukin-6 causes endothelial barrier dysfunction via the protein kinase C pathway. J. Surg. Res. 2002, 104, 118–123. [Google Scholar] [CrossRef] [PubMed]
  150. Förster, C.; Burek, M.; Romero, I.A.; Weksler, B.; Couraud, P.O.; Drenckhahn, D. Differential effects of hydrocortisone and TNFalpha on tight junction proteins in an in vitro model of the human blood-brain barrier. J. Physiol. 2008, 586, 1937–1949. [Google Scholar] [CrossRef]
  151. Stamatovic, S.M.; Keep, R.F.; Wang, M.M.; Jankovic, I.; Andjelkovic, A.V. Caveolae-mediated internalization of occludin and claudin-5 during CCL2-induced tight junction remodeling in brain endothelial cells. J. Biol. Chem. 2009, 284, 19053–19066. [Google Scholar] [CrossRef]
  152. Nagyoszi, P.; Wilhelm, I.; Farkas, A.E.; Fazakas, C.; Dung, N.T.K.; Haskó, J.; Krizbai, I.A. Expression and regulation of Toll-like receptors in cerebral endothelial cells. Neurochem. Int. 2010, 57, 556–564. [Google Scholar] [CrossRef]
  153. Racke, M.K.; Drew, P.D. Toll-like receptors in multiple sclerosis. Curr. Top. Microbiol. Immunol. 2009, 336, 155–168. [Google Scholar] [CrossRef]
  154. Sheikh, M.H.; Henson, S.M.; Loiola, R.A.; Mercurio, S.; Colamatteo, A.; Maniscalco, G.T.; De Rosa, V.; McArthur, S.; Solito, E. Immuno-metabolic impact of the multiple sclerosis patients’ sera on endothelial cells of the blood-brain barrier. J. Neuroinflamm. 2020, 17, 153. [Google Scholar] [CrossRef] [PubMed]
  155. Wittmann, G.; Mohácsik, P.; Balkhi, M.Y.; Gereben, B.; Lechan, R.M. Endotoxin-induced inflammation down-regulates l-type amino acid transporter 1 (LAT1) expression at the blood–brain barrier of male rats and mice. Fluids Barriers CNS 2015, 12, 21. [Google Scholar] [CrossRef]
  156. Roberts, D.J.; Goralski, K.B. A Critical Overview of the Influence of inflammation and infection on p-glycoprotein expression and activity in the brain. Expert Opin. Drug Metab. Toxicol. 2008, 4, 1245–1264. [Google Scholar] [CrossRef] [PubMed]
  157. Gelati, M.; Corsini, E.; Dufour, A.; Massa, G.; Giombini, S.; Solero, C.L.; Salmaggi, A. High-dose methylprednisolone reduces cytokine-induced adhesion molecules on human brain endothelium. Can. J. Neurol. Sci. 2000, 27, 241–244. [Google Scholar] [CrossRef] [PubMed]
  158. Ge, S.; Jiang, X.; Paul, D.; Song, L.; Wang, X.; Pachter, J.S. Human ES-derived MSCs correct TNF-α-mediated alterations in a blood–brain barrier model. Fluids Barriers CNS 2019, 16, 18. [Google Scholar] [CrossRef] [PubMed]
  159. Hou, Y.; Heon Ryu, C.; Jun, J.A.; Kim, S.M.; Jeong, C.H.; Jeun, S.-S. Interferon β-secreting mesenchymal stem cells combined with minocycline attenuate experimental autoimmune encephalomyelitis. J. Neuroimmunol. 2014, 274, 20–27. [Google Scholar] [CrossRef]
  160. Liu, Y.; Ma, Y.; Du, B.; Wang, Y.; Yang, G.-Y.; Bi, X. Mesenchymal stem cells attenuated blood-brain barrier disruption via downregulation of aquaporin-4 expression in EAE mice. Mol. Neurobiol. 2020, 57, 3891–3901. [Google Scholar] [CrossRef]
  161. Feigin, V.L.; Brainin, M.; Norrving, B.; Martins, S.; Sacco, R.L.; Hacke, W.; Fisher, M.; Pandian, J.; Lindsay, P. World Stroke Organization (WSO): Global stroke fact sheet 2022. Int. J. Stroke 2022, 17, 18–29. [Google Scholar] [CrossRef]
  162. Peng, L.; Hu, G.; Yao, Q.; Wu, J.; He, Z.; Law, B.Y.-K.; Hu, G.; Zhou, X.; Du, J.; Wu, A.; et al. Microglia autophagy in ischemic stroke: A double-edged sword. Front. Immunol. 2022, 13, 1013311. [Google Scholar] [CrossRef]
  163. Iadecola, C.; Anrather, J. The immunology of stroke: From mechanisms to translation. Nat. Med. 2011, 17, 796–808. [Google Scholar] [CrossRef]
  164. Bernardo-Castro, S.; Sousa, J.A.; Brás, A.; Cecília, C.; Rodrigues, B.; Almendra, L.; Machado, C.; Santo, G.; Silva, F.; Ferreira, L.; et al. Pathophysiology of blood-brain barrier permeability throughout the different stages of ischemic stroke and its implication on hemorrhagic transformation and recovery. Front. Neurol. 2020, 11, 594672. [Google Scholar] [CrossRef] [PubMed]
  165. Magee, P.N.; Stoner, H.B.; Barnes, J.M. The experimental production in oedema in the central nervous system of the rat by triethyltin compounds. J. Pathol. 1957, 73, 107–124. [Google Scholar] [CrossRef]
  166. Liebeskind, D.S.; Jüttler, E.; Shapovalov, Y.; Yegin, A.; Landen, J.; Jauch, E.C. Cerebral edema associated with large hemispheric infarction. Stroke 2019, 50, 2619–2625. [Google Scholar] [CrossRef]
  167. Yang, Y.; Rosenberg, G.A. Blood-brain barrier breakdown in acute and chronic cerebrovascular disease. Stroke 2011, 42, 3323–3328. [Google Scholar] [CrossRef] [PubMed]
  168. Price, C.J.; Menon, D.K.; Peters, A.M.; Ballinger, J.R.; Barber, R.W.; Balan, K.K.; Lynch, A.; Xuereb, J.H.; Fryer, T.; Guadagno, J.V.; et al. Cerebral neutrophil recruitment, histology, and outcome in acute ischemic stroke: An imaging-based study. Stroke 2004, 35, 1659–1664. [Google Scholar] [CrossRef] [PubMed]
  169. Krizbai, I.A.; Bauer, H.; Bresgen, N.; Eckl, P.M.; Farkas, A.; Szatmári, E.; Traweger, A.; Wejksza, K.; Bauer, H.-C. Effect of oxidative stress on the junctional proteins of cultured cerebral endothelial cells. Cell Mol. Neurobiol. 2005, 25, 129–139. [Google Scholar] [CrossRef]
  170. Turner, R.J.; Sharp, F.R. Implications of MMP9 for blood brain barrier disruption and hemorrhagic transformation following ischemic stroke. Front. Cell Neurosci. 2016, 10, 56. [Google Scholar] [CrossRef]
  171. Qiu, Y.M.; Zhang, C.L.; Chen, A.Q.; Wang, H.L.; Zhou, Y.F.; Li, Y.N.; Hu, B. Immune cells in the BBB disruption after acute ischemic stroke: Targets for immune therapy? Front. Immunol. 2021, 12, 678744. [Google Scholar] [CrossRef]
  172. Kang, L.; Yu, H.; Yang, X.; Zhu, Y.; Bai, X.; Wang, R.; Cao, Y.; Xu, H.; Luo, H.; Lu, L.; et al. Neutrophil extracellular traps released by neutrophils impair revascularization and vascular remodeling after stroke. Nat. Commun. 2020, 11, 2488. [Google Scholar] [CrossRef]
  173. Jickling, G.C.; Liu, D.; Ander, B.P.; Stamova, B.; Zhan, X.; Sharp, F.R. Targeting neutrophils in ischemic stroke: Translational insights from experimental studies. J. Cereb. Blood Flow. Metab. 2015, 35, 888–901. [Google Scholar] [CrossRef]
  174. Kataoka, H.; Kim, S.W.; Plesnila, N. Leukocyte-endothelium interactions during permanent focal cerebral ischemia in mice. J. Cereb. Blood Flow. Metab. 2004, 24, 668–676. [Google Scholar] [CrossRef] [PubMed]
  175. Tsai, N.W.; Chang, W.N.; Shaw, C.F.; Jan, C.R.; Huang, C.R.; Chen, S.D.; Chuang, Y.C.; Lee, L.H.; Lu, C.H. The value of leukocyte adhesion molecules in patients after ischemic stroke. J. Neurol. 2009, 256, 1296–1302. [Google Scholar] [CrossRef] [PubMed]
  176. Gan, Y.; Liu, Q.; Wu, W.; Yin, J.-X.; Bai, X.-F.; Shen, R.; Wang, Y.; Chen, J.; La Cava, A.; Poursine-Laurent, J.; et al. Ischemic neurons recruit natural killer cells that accelerate brain infarction. Proc. Natl. Acad. Sci. USA 2014, 111, 2704–2709. [Google Scholar] [CrossRef] [PubMed]
  177. Kurzepa, J.; Kurzepa, J.; Golab, P.; Czerska, S.; Bielewicz, J. The significance of matrix metalloproteinase (MMP)-2 and MMP-9 in the ischemic stroke. Int. J. Neurosci. 2014, 124, 707–716. [Google Scholar] [CrossRef] [PubMed]
  178. Lee, C.W.; Stabile, E.; Kinnaird, T.; Shou, M.; Devaney, J.M.; Epstein, S.E.; Burnett, M.S. Temporal patterns of gene expression after acute hindlimb ischemia in mice: Insights into the genomic program for collateral vessel development. J. Am. Coll. Cardiol. 2004, 43, 474–482. [Google Scholar] [CrossRef]
  179. Liu, R.; Pan, M.-X.; Tang, J.-C.; Zhang, Y.; Liao, H.-B.; Zhuang, Y.; Zhao, D.; Wan, Q. Role of neuroinflammation in ischemic stroke. Neurol. Neuroimmunol. Neuroinflamm 2017, 4, 158–166. [Google Scholar] [CrossRef]
  180. Krupinski, J.; Kaluza, J.; Kumar, P.; Kumar, S.; Wang, J.M. Role of angiogenesis in patients with cerebral ischemic stroke. Stroke 1994, 25, 1794–1798. [Google Scholar] [CrossRef]
  181. Navarro-Sobrino, M.; Rosell, A.; Hernández-Guillamon, M.; Penalba, A.; Boada, C.; Domingues-Montanari, S.; Ribó, M.; Alvarez-Sabín, J.; Montaner, J. A large screening of angiogenesis biomarkers and their association with neurological outcome after ischemic stroke. Atherosclerosis 2011, 216, 205–211. [Google Scholar] [CrossRef]
  182. Jiang, X.; Andjelkovic, A.V.; Zhu, L.; Yang, T.; Bennett, M.V.L.; Chen, J.; Keep, R.F.; Shi, Y. Blood-brain barrier dysfunction and recovery after ischemic stroke. Prog. Neurobiol. 2018, 163–164, 144–171. [Google Scholar] [CrossRef]
  183. Shindo, A.; Maki, T.; Mandeville, E.T.; Liang, A.C.; Egawa, N.; Itoh, K.; Itoh, N.; Borlongan, M.; Holder, J.C.; Chuang, T.T.; et al. Astrocyte-derived pentraxin 3 supports blood-brain barrier integrity under acute phase of stroke. Stroke 2016, 47, 1094–1100. [Google Scholar] [CrossRef]
  184. Lin, R.; Cai, J.; Nathan, C.; Wei, X.; Schleidt, S.; Rosenwasser, R.; Iacovitti, L. Neurogenesis is enhanced by stroke in multiple new stem cell niches along the ventricular system at sites of high BBB permeability. Neurobiol. Dis. 2015, 74, 229–239. [Google Scholar] [CrossRef] [PubMed]
  185. Zhang, R.L.; Chopp, M.; Roberts, C.; Liu, X.; Wei, M.; Nejad-Davarani, S.P.; Wang, X.; Zhang, Z.G. Stroke increases neural stem cells and angiogenesis in the neurogenic niche of the adult mouse. PLoS ONE 2014, 9, e113972. [Google Scholar] [CrossRef]
  186. Do, P.T.; Wu, C.-C.; Chiang, Y.-H.; Hu, C.-J.; Chen, K.-Y. Mesenchymal stem/stromal cell therapy in blood–brain barrier preservation following ischemia: Molecular mechanisms and prospects. Int. J. Mol. Sci. 2021, 22, 10045. [Google Scholar] [CrossRef]
  187. Huang, Y.; Wang, J.; Cai, J.; Qiu, Y.; Zheng, H.; Lai, X.; Sui, X.; Wang, Y.; Lu, Q.; Zhang, Y.; et al. Targeted homing of CCR2-overexpressing mesenchymal stromal cells to ischemic brain enhances post-stroke recovery partially through PRDX4-mediated blood-brain barrier preservation. Theranostics 2018, 8, 5929–5944. [Google Scholar] [CrossRef]
  188. Cheng, Z.; Wang, L.; Qu, M.; Liang, H.; Li, W.; Li, Y.; Deng, L.; Zhang, Z.; Yang, G.-Y. Mesenchymal stem cells attenuate blood-brain barrier leakage after cerebral ischemia in mice. J. Neuroinflamm. 2018, 15, 135. [Google Scholar] [CrossRef]
  189. Liu, K.; Ji, K.; Guo, L.; Wu, W.; Lu, H.; Shan, P.; Yan, C. Mesenchymal stem cells rescue injured endothelial cells in an in vitro ischemia–reperfusion model via tunneling nanotube like structure-mediated mitochondrial transfer. Microvasc. Res. 2014, 92, 10–18. [Google Scholar] [CrossRef] [PubMed]
  190. Liu, K.; Guo, L.; Zhou, Z.; Pan, M.; Yan, C. Mesenchymal stem cells transfer mitochondria into cerebral microvasculature and promote recovery from ischemic stroke. Microvasc. Res. 2019, 123, 74–80. [Google Scholar] [CrossRef] [PubMed]
  191. Zacharek, A.; Chen, J.; Li, A.; Cui, X.; Li, Y.; Roberts, C.; Feng, Y.; Gao, Q.; Chopp, M. Angiopoietin1/TIE2 and VEGF/FLK1 induced by MSC treatment amplifies angiogenesis and vascular stabilization after stroke. J. Cereb. Blood Flow. Metab. 2007, 27, 1684–1691. [Google Scholar] [CrossRef]
  192. Gregorius, J.; Wang, C.; Stambouli, O.; Hussner, T.; Qi, Y.; Tertel, T.; Börger, V.; Mohamud Yusuf, A.; Hagemann, N.; Yin, D.; et al. Small extracellular vesicles obtained from hypoxic mesenchymal stromal cells have unique characteristics that promote cerebral angiogenesis, brain remodeling and neurological recovery after focal cerebral ischemia in mice. Basic. Res. Cardiol. 2021, 116, 40. [Google Scholar] [CrossRef]
  193. Hu, H.; Hu, X.; Li, L.; Fang, Y.; Yang, Y.; Gu, J.; Xu, J.; Chu, L. Exosomes derived from bone marrow mesenchymal stem cells promote angiogenesis in ischemic stroke mice via upregulation of miR-21-5p. Biomolecules 2022, 12, 883. [Google Scholar] [CrossRef]
  194. Xia, Y.; Ling, X.; Hu, G.; Zhu, Q.; Zhang, J.; Li, Q.; Zhao, B.; Wang, Y.; Deng, Z. Small extracellular vesicles secreted by human iPSC-derived MSC enhance angiogenesis through inhibiting STAT3-dependent autophagy in ischemic stroke. Stem Cell Res. Ther. 2020, 11, 313. [Google Scholar] [CrossRef] [PubMed]
  195. Kvistad, C.E.; Kråkenes, T.; Gjerde, C.; Mustafa, K.; Rekand, T.; Bø, L. Safety and clinical efficacy of mesenchymal stem cell treatment in traumatic spinal cord injury, multiple sclerosis and ischemic stroke—A systematic review and meta-analysis. Front. Neurol. 2022, 13, 891514. [Google Scholar] [CrossRef] [PubMed]
  196. Regmi, S.; Liu, D.D.; Shen, M.; Kevadiya, B.D.; Ganguly, A.; Primavera, R.; Chetty, S.; Yarani, R.; Thakor, A.S. Mesenchymal stromal cells for the treatment of Alzheimer’s disease: Strategies and Limitations. Front. Mol. Neurosci. 2022, 15, 1011225. [Google Scholar] [CrossRef] [PubMed]
  197. Galipeau, J.; Sensébé, L. Mesenchymal stromal cells: Clinical challenges and therapeutic opportunities. Cell Stem Cell 2018, 22, 824–833. [Google Scholar] [CrossRef]
  198. Fričová, D.; Korchak, J.A.; Zubair, A.C. Challenges and translational considerations of mesenchymal stem/stromal cell therapy for Parkinson’s disease. NPJ Regen. Med. 2020, 5, 20. [Google Scholar] [CrossRef]
  199. Borlongan, C.V. Concise Review: Stem Cell Therapy for Stroke Patients: Are We There Yet? Stem Cells Transl. Med. 2019, 8, 983–988. [Google Scholar] [CrossRef]
  200. Yousefi, F.; Lavi Arab, F.; Saeidi, K.; Amiri, H.; Mahmoudi, M. Various strategies to improve efficacy of stem cell transplantation in multiple sclerosis: Focus on mesenchymal stem cells and neuroprotection. J. Neuroimmunol. 2019, 328, 20–34. [Google Scholar] [CrossRef]
  201. Russell, A.L.; Lefavor, R.; Durand, N.; Glover, L.; Zubair, A.C. Modifiers of mesenchymal stem cell quantity and quality. Transfusion 2018, 58, 1434–1440. [Google Scholar] [CrossRef]
  202. Kwon, H.M.; Hur, S.M.; Park, K.Y.; Kim, C.K.; Kim, Y.M.; Kim, H.S.; Shin, H.C.; Won, M.H.; Ha, K.S.; Kwon, Y.G.; et al. Multiple paracrine factors secreted by mesenchymal stem cells contribute to angiogenesis. Vasc. Pharm. 2014, 63, 19–28. [Google Scholar] [CrossRef]
  203. Wilson, A.; Butler, P.E.; Seifalian, A.M. Adipose-derived stem cells for clinical applications: A review. Cell Prolif. 2011, 44, 86–98. [Google Scholar] [CrossRef]
  204. Bowen, J.E. Technical issues in harvesting and concentrating stem cells (bone marrow and adipose). PMR 2015, 7, S8–S18. [Google Scholar] [CrossRef] [PubMed]
  205. Pittenger, M.F.; Mackay, A.M.; Beck, S.C.; Jaiswal, R.K.; Douglas, R.; Mosca, J.D.; Moorman, M.A.; Simonetti, D.W.; Craig, S.; Marshak, D.R. Multilineage potential of adult human mesenchymal stem cells. Science 1999, 284, 143–147. [Google Scholar] [CrossRef] [PubMed]
  206. Mastrolia, I.; Foppiani, E.M.; Murgia, A.; Candini, O.; Samarelli, A.V.; Grisendi, G.; Veronesi, E.; Horwitz, E.M.; Dominici, M. Challenges in clinical development of mesenchymal stromal/stem cells: Concise review. Stem Cells Transl. Med. 2019, 8, 1135–1148. [Google Scholar] [CrossRef] [PubMed]
  207. Marquez-Curtis, L.A.; Janowska-Wieczorek, A.; McGann, L.E.; Elliott, J.A. Mesenchymal stromal cells derived from various tissues: Biological, clinical and cryopreservation aspects. Cryobiology 2015, 71, 181–197. [Google Scholar] [CrossRef]
  208. Moll, G.; Geissler, S.; Catar, R.; Ignatowicz, L.; Hoogduijn, M.J.; Strunk, D.; Bieback, K.; Ringden, O. Cryopreserved or Fresh Mesenchymal Stromal Cells: Only a Matter of Taste or Key to Unleash the Full Clinical Potential of MSC Therapy? Adv. Exp. Med. Biol. 2016, 951, 77–98. [Google Scholar] [CrossRef]
  209. Efimenko, A.Y.; Kochegura, T.N.; Akopyan, Z.A.; Parfyonova, Y.V. Autologous stem cell therapy: How aging and chronic diseases affect stem and progenitor cells. Biores Open Access 2015, 4, 26–38. [Google Scholar] [CrossRef]
  210. Levy, M.L.; Crawford, J.R.; Dib, N.; Verkh, L.; Tankovich, N.; Cramer, S.C. Phase I/II study of safety and preliminary efficacy of intravenous allogeneic mesenchymal stem cells in chronicstroke. Stroke 2019, 50, 2835–2841. [Google Scholar] [CrossRef]
  211. Berglund, A.K.; Fortier, L.A.; Antczak, D.F.; Schnabel, L.V. Immunoprivileged no more: Measuring the immunogenicity of allogeneic adult mesenchymal stem cells. Stem Cell Res. Ther. 2017, 8, 288. [Google Scholar] [CrossRef]
  212. Li, Y.; Lin, F. Mesenchymal stem cells are injured by complement after their contact with serum. Blood. 2012, 120, 3436–3443. [Google Scholar] [CrossRef]
  213. Nauta, A.J.; Westerhuis, G.; Kruisselbrink, A.B.; Lurvink, E.G.; Willemze, R.; Fibbe, W.E. Donor-derived mesenchymal stem cells are immunogenic in an allogeneic host and stimulate donor graft rejection in a nonmyeloablative setting. Blood 2006, 108, 2114–2120. [Google Scholar] [CrossRef]
  214. Diamandis, T.; Borlongan, C.V. One, two, three steps toward cell therapy for stroke. Stroke 2015, 46, 588–591. [Google Scholar] [CrossRef] [PubMed]
  215. Bang, O.Y.; Lee, J.S.; Lee, P.H.; Lee, G. Autologous mesenchymal stem cell transplantation in stroke patients. Ann. Neurol. 2005, 57, 874–882. [Google Scholar] [CrossRef] [PubMed]
  216. Prasad, K.; Sharma, A.; Garg, A.; Mohanty, S.; Bhatnagar, S.; Johri, S.; Singh, K.K.; Nair, V.; Sarkar, R.S.; Gorthi, S.P.; et al. Intravenous autologous bone marrow mononuclear stem cell therapy for ischemic stroke: A multicentric, randomized trial. Stroke 2014, 45, 3618–3624. [Google Scholar] [CrossRef] [PubMed]
  217. Banerjee, S.; Bentley, P.; Hamady, M.; Marley, S.; Davis, J.; Shlebak, A.; Nicholls, J.; Williamson, D.A.; Jensen, S.L.; Gordon, M.; et al. Intra-Arterial Immunoselected CD34+ Stem Cells for Acute Ischemic Stroke. Stem Cells Transl. Med. 2014, 3, 1322–1330. [Google Scholar] [CrossRef]
  218. Savitz, S.I.; Misra, V.; Kasam, M.; Juneja, H.; Cox, C.S., Jr.; Alderman, S.; Aisiku, I.; Kar, S.; Gee, A.; Grotta, J.C. Intravenous autologous bone marrow mononuclear cells for ischemic stroke. Ann. Neurol. 2011, 70, 59–69. [Google Scholar] [CrossRef]
  219. Liu, L.; Eckert, M.A.; Riazifar, H.; Kang, D.K.; Agalliu, D.; Zhao, W. From blood to the brain: Can systemically transplanted mesenchymal stem cells cross the blood-brain barrier? Stem Cells Int. 2013, 2013, 435093. [Google Scholar] [CrossRef]
  220. Ruigrok, M.J.; de Lange, E.C. Emerging insights for translational pharmacokinetic and pharmacokinetic-pharmacodynamic studies: Towards prediction of nose-to-brain transport in humans. AAPS J. 2015, 17, 493–505. [Google Scholar] [CrossRef]
  221. Feng, C.J.; Perng, C.K.; Lin, C.H.; Tsai, C.H.; Huang, P.H.; Ma, H. Intra-Arterial Injection of Human Adipose-Derived Stem Cells Improves Viability of the Random Component of Axial Skin Flaps in Nude Mice. J. Plast. Reconstr. Aesthet. Surg. 2020, 73, 598–607. [Google Scholar] [CrossRef]
  222. Lundberg, J.; Jussing, E.; Liu, Z.; Meng, Q.; Rao, M.; Samén, E.; Grankvist, R.; Damberg, P.; Dodoo, E.; Maeurer, M.; et al. Safety of Intra-Arterial Injection with Tumor-Activated T Cells to the Rabbit Brain Evaluated by MRI and SPECT/CT. Cell Transplant. 2017, 26, 283–292. [Google Scholar] [CrossRef]
  223. Tatsumi, K.; Ohashi, K.; Matsubara, Y.; Kohori, A.; Ohno, T.; Kakidachi, H.; Horii, A.; Kanegae, K.; Utoh, R.; Iwata, T.; et al. Tissue factor triggers procoagulation in transplanted mesenchymal stem cells leading to thromboembolism. Biochem. Biophys. Res. Commun. 2013, 431, 203–209. [Google Scholar] [CrossRef]
  224. Liu, S.; Zhou, J.; Zhang, X.; Liu, Y.; Chen, J.; Hu, B.; Song, J.; Zhang, Y. Strategies to Optimize Adult Stem Cell Therapy for Tissue Regeneration. Int. J. Mol. Sci. 2016, 17, 982. [Google Scholar] [CrossRef]
  225. Payne, N.; Siatskas, C.; Bernard, C.C. The promise of stem cell and regenerative therapies for multiple sclerosis. J. Autoimmun. 2008, 31, 288–294. [Google Scholar] [CrossRef]
  226. Karussis, D.; Karageorgiou, C.; Vaknin-Dembinsky, A.; Gowda-Kurkalli, B.; Gomori, J.M.; Kassis, I.; Bulte, J.W.; Petrou, P.; Ben-Hur, T.; Abramsky, O.; et al. Safety and immunological effects of mesenchymal stem cell transplantation in patients with multiple sclerosis and amyotrophic lateral sclerosis. Arch. Neurol. 2010, 67, 1187–1194. [Google Scholar] [CrossRef] [PubMed]
  227. Uccelli, A.; Laroni, A.; Ali, R.; Battaglia, M.A.; Blinkenberg, M.; Brundin, L.; Clanet, M.; Fernandez, O.; Marriot, J.; Muraro, P.; et al. MESEMS investigators. Safety, tolerability, and activity of mesenchymal stem cells versus placebo in multiple sclerosis (MESEMS): A phase 2, randomised, double-blind crossover trial. Lancet Neurol. 2021, 20, 917–929. [Google Scholar] [CrossRef] [PubMed]
  228. Zhou, Y.; Zhang, X.; Xue, H.; Liu, L.; Zhu, J.; Jin, T. Autologous mesenchymal stem cell transplantation in multiple sclerosis: A metaanalysis. Stem Cells Int. 2019, 2019, 8536785. [Google Scholar] [CrossRef] [PubMed]
  229. Hanson, L.R.; Frey, W.H., II. Intranasal delivery bypasses the blood-brain barrier to target therapeutic agents to the central nervous system and treat neurodegenerative disease. BMC Neurosci. 2008, 9, S5. [Google Scholar] [CrossRef]
  230. Danielyan, L.; Beer-Hammer, S.; Stolzing, A.; Schäfer, R.; Siegel, G.; Fabian, C.; Kahle, P.; Biedermann, T.; Lourhmati, A.; Buadze, M.; et al. Intranasal delivery of bone marrow-derived mesenchymal stem cells, macrophages, and microglia to the brain in mouse models of Alzheimer’s and Parkinson’s disease. Cell Transpl. 2014, 23, S123–S139. [Google Scholar] [CrossRef]
  231. Salama, M.; Sobh, M.; Emam, M.; Abdalla, A.; Sabry, D.; El-Gamal, M.; Lotfy, A.; El-Husseiny, M.; Sobh, M.; Shalash, A.; et al. Effect of intranasal stem cell administration on the nigrostriatal system in a mouse model of Parkinson’s disease. Exp. Ther. Med. 2017, 13, 976–982. [Google Scholar] [CrossRef]
  232. Lavoie, J.R.; Rosu-Myles, M. Uncovering the secretes of mesenchymal stem cells. Biochimie 2013, 95, 2212–2221. [Google Scholar] [CrossRef]
  233. Kim, S.J.; Moon, G.J.; Chang, W.H.; Kim, Y.H.; Bang, O.Y. Intravenous transplantation of mesenchymal stem cells preconditioned with early phase stroke serum: Current evidence and study protocol for a randomized trial. Trials 2013, 14, 317. [Google Scholar] [CrossRef]
  234. Oshita, J.; Okazaki, T.; Mitsuhara, T.; Imura, T.; Nakagawa, K.; Otsuka, T.; Kurose, T.; Tamura, T.; Abiko, M.; Takeda, M.; et al. Early transplantation of human cranial bone-derived mesenchymal stem cells enhances functional recovery in ischemic stroke model rats. Neurol. Med. Chir. 2020, 60, 83–93. [Google Scholar] [CrossRef] [PubMed]
  235. Jennings, A.; Carroll, W. Quantification of oligodendrocyte progenitor cells in human and cat optic nerve: Implications for endogenous repair in multiple sclerosis. Glia 2010, 58, 1425–1436. [Google Scholar] [CrossRef] [PubMed]
  236. Miller, R.H.; Bai, L. Translating stem cell therapies to the clinic. Neurosci. Lett. 2012, 519, 87–92. [Google Scholar] [CrossRef] [PubMed]
  237. Hess, D.C.; Wechsler, L.R.; Clark, W.M.; Savitz, S.I.; Ford, G.A.; Chiu, D.; Yavagal, D.R.; Uchino, K.; Liebeskind, D.S.; Auchus, A.P.; et al. Safety and efficacy of multipotent adult progenitor cells in acute ischaemic stroke (MASTERS): A randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Neurol. 2017, 16, 360–368. [Google Scholar] [CrossRef]
  238. Steinberg, G.K.; Kondziolka, D.; Wechsler, L.R.; Lunsford, L.D.; Kim, A.S.; Johnson, J.N.; Bates, D.; Poggio, G.; Case, C.; McGrogan, M.; et al. Two-year safety and clinical outcomes in chronic ischemic stroke patients after implantation of modified bone marrow-derived mesenchymal stem cells (SB623): A phase 1/2a study. J. Neurosurg. 2018, 131, 1462–1472. [Google Scholar] [CrossRef]
  239. Steinberg, G.K.; Kondziolka, D.; Wechsler, L.R.; Lunsford, L.D.; Coburn, M.L.; Billigen, J.B.; Kim, A.S.; Johnson, J.N.; Bates, D.; King, B.; et al. Clinical Outcomes of Transplanted Modified Bone Marrow-Derived Mesenchymal Stem Cells in Stroke: A Phase 1/2a Study. Stroke 2016, 47, 1817–1824. [Google Scholar] [CrossRef]
  240. Prange, S.; Metereau, E.; Thobois, S. Structural imaging in Parkinson’s disease: New developments. Curr. Neurol. Neurosci. Rep. 2017, 19, 50. [Google Scholar] [CrossRef]
  241. Jimenez-Jimenez, F.J.; Alonso-Navarro, H.; Garcia-Martin, E.; Agundez, J.A. Cerebrospinal fluid biochemical studies in patients with Parkinson’s disease: Toward a potential search for biomarkers for this disease. Front. Cell Neurosci. 2014, 8, 36. [Google Scholar] [CrossRef]
  242. Le Blanc, K.; Mougiakakos, D. Multipotent mesenchymal stromal cells and the innate immune system. Nat. Rev. Immunol. 2012, 12, 383–396. [Google Scholar] [CrossRef]
  243. Yin, J.Q.; Zhu, J.; Ankrum, J.A. Manufacturing of primed mesenchymal stromal cells for therapy. Nat. Biomed. Eng. 2019, 3, 90–104. [Google Scholar] [CrossRef]
  244. Moon, G.J.; Sung, J.H.; Kim, D.H.; Kim, E.H.; Cho, Y.H.; Son, J.P.; Cha, J.M.; Bang, O.Y. Application of Mesenchymal Stem Cell-Derived Extracellular Vesicles for Stroke: Biodistribution and MicroRNA Study. Transl. Stroke Res. 2019, 10, 509–521. [Google Scholar] [CrossRef] [PubMed]
  245. Grange, C.; Tapparo, M.; Bruno, S.; Chatterjee, D.; Quesenberry, P.J.; Tetta, C.; Camussi, G. Biodistribution of mesenchymal stem cell-derived extracellular vesicles in a model of acute kidney injury monitored by optical imaging. Int. J. Mol. Med. 2014, 33, 1055–1063. [Google Scholar] [CrossRef] [PubMed]
  246. Kordelas, L.; Rebmann, V.; Ludwig, A.K.; Radtke, S.; Ruesing, J.; Doeppner, T.R.; Epple, M.; Horn, P.A.; Beelen, D.W.; Giebel, B. MSC-derived exosomes: A novel tool to treat therapy-refractory graft-versus-host disease. Leukemia 2014, 28, 970–973. [Google Scholar] [CrossRef] [PubMed]
  247. Katagiri, W.; Osugi, M.; Kawai, T.; Hibi, H. First-in-human study and clinical case reports of the alveolar bone regeneration with the secretome from human mesenchymal stem cells. Head. Face Med. 2016, 12, 5. [Google Scholar] [CrossRef]
  248. Zhang, X.; Liu, J.; Yu, B.; Ma, F.; Ren, X.; Li, X. Effects of mesenchymal stem cells and their exosomes on the healing of large and refractory macular holes. Graefes Arch. Clin. Exp. Ophthalmol. 2018, 256, 2041–2052. [Google Scholar] [CrossRef]
  249. Fafián-Labora, J.; Lesende-Rodriguez, I.; Fernández-Pernas, P.; Sangiao-Alvarellos, S.; Monserrat, L.; Arntz, O.J.; van de Loo, F.J.; Mateos, J.; Arufe, M.C. Effect of age on pro-inflammatory miRNAs contained in mesenchymal stem cell-derived extracellular vesicles. Sci. Rep. 2017, 7, 43923. [Google Scholar] [CrossRef]
  250. Cash, A.; Theus, M.H. Mechanisms of Blood-Brain Barrier Dysfunction in Traumatic Brain Injury. Int. J. Mol. Sci. 2020, 21, 3344. [Google Scholar] [CrossRef]
  251. Sulhan, S.; Lyon, K.A.; Shapiro, L.A.; Huang, J.H. Neuroinflammation and blood-brain barrier disruption following traumatic brain injury: Pathophysiology and potential therapeutic targets. J. Neurosci. Res. 2020, 98, 19–28. [Google Scholar] [CrossRef]
  252. Jin, L.Y.; Li, J.; Wang, K.F.; Xia, W.W.; Zhu, Z.Q.; Wang, C.R.; Li, X.F.; Liu, H.Y. Blood-Spinal Cord Barrier in Spinal Cord Injury: A Review. J. Neurotrauma. 2021, 38, 1203–1224. [Google Scholar] [CrossRef]
  253. Kakaroubas, N.; Brennan, S.; Keon, M.; Saksena, N.K. Pathomechanisms of Blood-Brain Barrier Disruption in ALS. Neurosci. J. 2019, 2019, 2537698. [Google Scholar] [CrossRef]
  254. Alarcan, H.; Al Ojaimi, Y.; Lanznaster, D.; Escoffre, J.M.; Corcia, P.; Vourc’h, P.; Andres, C.R.; Veyrat-Durebex, C.; Blasco, H. Taking Advantages of Blood-Brain or Spinal Cord Barrier Alterations or Restoring Them to Optimize Therapy in ALS? J. Pers. Med. 2022, 12, 1071. [Google Scholar] [CrossRef] [PubMed]
  255. Sweeney, M.D.; Sagare, A.P.; Zlokovic, B.V. Blood-brain barrier breakdown in Alzheimer disease and other neurodegenerative disorders. Nat. Rev. Neurol. 2018, 14, 133–150. [Google Scholar] [CrossRef] [PubMed]
  256. Dadas, A.; Janigro, D. Breakdown of blood brain barrier as a mechanism of post-traumatic epilepsy. Neurobiol. Dis. 2019, 123, 20–26. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of the blood-brain barrier (BBB). It is located within the neurovascular unit (NVU) and constituted by endothelial cells interconnected by tight junctions (TJ) and adherens junctions (AJs), and neighboring cells, such as pericytes, astrocytes, neurons, and microglia. CNS: central nervous system; BMVECs: brain microvascular endothelial cells; ZO: zonula occludens; PECAM: Platelet endothelial cell adhesion molecule; VE-cadherin: vascular endothelial cadherin.
Figure 1. Schematic representation of the blood-brain barrier (BBB). It is located within the neurovascular unit (NVU) and constituted by endothelial cells interconnected by tight junctions (TJ) and adherens junctions (AJs), and neighboring cells, such as pericytes, astrocytes, neurons, and microglia. CNS: central nervous system; BMVECs: brain microvascular endothelial cells; ZO: zonula occludens; PECAM: Platelet endothelial cell adhesion molecule; VE-cadherin: vascular endothelial cadherin.
Ijms 24 14117 g001
Figure 2. Blood-brain barrier (BBB) disruption in Alzheimer’s disease (AD) pathogenesis. The breakdown of the BBB in AD is characterized by a cascade of events, including loss of TJ integrity, disorganization of the basement membrane (BM) and extracellular matrix, pericyte degeneration and detachment, activation of glial cells, astrocyte depolarization, and alteration of BBB transporter expression (LRP1, P-gp, and GLUT1 are reduced, whereas RAGE expression is increased). These modifications may lead directly or indirectly to disturbed Amyloid β (Aβ) clearance in the neurovascular unit (NVU), contributing to further neuronal toxicity and AD pathogenesis. Treatment with MSCs restore BBB integrity by stabilizing TJ, BM, and extracellular matrix (ECM) remodeling, and reduces the neuroinflammation and Aβ accumulation. CNS: central nervous system; BMVECs: brain microvascular endothelial cells; BM: basement membrane; RAGE: receptor for advanced glycation end products; LRP1: Low density lipoprotein receptor-related protein 1; P-gp: permeability glycoprotein; GLUT1: glucose transporter 1; MMP: metalloproteinases; TJ: tight junctions; AQP: aquaporin. Blue and red arrows mean increase and reduction, respectively.
Figure 2. Blood-brain barrier (BBB) disruption in Alzheimer’s disease (AD) pathogenesis. The breakdown of the BBB in AD is characterized by a cascade of events, including loss of TJ integrity, disorganization of the basement membrane (BM) and extracellular matrix, pericyte degeneration and detachment, activation of glial cells, astrocyte depolarization, and alteration of BBB transporter expression (LRP1, P-gp, and GLUT1 are reduced, whereas RAGE expression is increased). These modifications may lead directly or indirectly to disturbed Amyloid β (Aβ) clearance in the neurovascular unit (NVU), contributing to further neuronal toxicity and AD pathogenesis. Treatment with MSCs restore BBB integrity by stabilizing TJ, BM, and extracellular matrix (ECM) remodeling, and reduces the neuroinflammation and Aβ accumulation. CNS: central nervous system; BMVECs: brain microvascular endothelial cells; BM: basement membrane; RAGE: receptor for advanced glycation end products; LRP1: Low density lipoprotein receptor-related protein 1; P-gp: permeability glycoprotein; GLUT1: glucose transporter 1; MMP: metalloproteinases; TJ: tight junctions; AQP: aquaporin. Blue and red arrows mean increase and reduction, respectively.
Ijms 24 14117 g002
Figure 3. Schematic image illustrating blood-brain barrier dysfunction in PD. Alterations in BMVECs are characterized by decreased TJ and AJ proteins and disorganization of the basement membrane. In addition to the reduction in astrocytic end-feet and pericyte loss, these changes can lead to BBB breakdown and subsequent accumulation of fibrinogen, thrombin, and hemosiderin, which together with α-synuclein can activate glial cells and injure dopaminergic neurons. Treatment with MSCs recovers BBB integrity by stabilizing the TJ structure and decreases the production and accumulation of neuroinflammatory and neurotoxic mediators. CNS: central nervous system; BMVECs: brain microvascular endothelial cells; TJ: tight junctions; P-gp: permeability glycoprotein; Ig: immunoglobulin. Blue and red arrows mean increase and reduction, respectively.
Figure 3. Schematic image illustrating blood-brain barrier dysfunction in PD. Alterations in BMVECs are characterized by decreased TJ and AJ proteins and disorganization of the basement membrane. In addition to the reduction in astrocytic end-feet and pericyte loss, these changes can lead to BBB breakdown and subsequent accumulation of fibrinogen, thrombin, and hemosiderin, which together with α-synuclein can activate glial cells and injure dopaminergic neurons. Treatment with MSCs recovers BBB integrity by stabilizing the TJ structure and decreases the production and accumulation of neuroinflammatory and neurotoxic mediators. CNS: central nervous system; BMVECs: brain microvascular endothelial cells; TJ: tight junctions; P-gp: permeability glycoprotein; Ig: immunoglobulin. Blue and red arrows mean increase and reduction, respectively.
Ijms 24 14117 g003
Figure 4. Blood-brain barrier alterations in multiple sclerosis. Hallmark features of MS development include an early BBB breakdown accompanied by reduced TJ expression, endothelial degeneration, leukocyte infiltration, and neuroimmune activation. Treatment with MSCs exerts immunomodulatory and neuroprotective roles in MS that in turn involve the stabilization of BBB integrity. CNS: central nervous system; ECM: extracellular matrix; BMVECs: brain microvascular endothelial cells; ECAMs: endothelial cell adhesion molecules; ICAM: intercellular adhesion molecules; TJ: tight junctions; TLR: Toll-like receptor; GLUT1: glucose transporter; Ig: immunoglobulin. Blue and red arrows mean increase and reduction, respectively.
Figure 4. Blood-brain barrier alterations in multiple sclerosis. Hallmark features of MS development include an early BBB breakdown accompanied by reduced TJ expression, endothelial degeneration, leukocyte infiltration, and neuroimmune activation. Treatment with MSCs exerts immunomodulatory and neuroprotective roles in MS that in turn involve the stabilization of BBB integrity. CNS: central nervous system; ECM: extracellular matrix; BMVECs: brain microvascular endothelial cells; ECAMs: endothelial cell adhesion molecules; ICAM: intercellular adhesion molecules; TJ: tight junctions; TLR: Toll-like receptor; GLUT1: glucose transporter; Ig: immunoglobulin. Blue and red arrows mean increase and reduction, respectively.
Ijms 24 14117 g004
Figure 5. Scheme illustrating pathophysiology of blood-brain barrier permeability during stroke. Left panel, hyperacute and acute stages are characterized by endothelial cell destabilization and BBB disruption, edema, peripheral immune infiltration, and glial immune activation. Right panel, subacute and chronic stages are involved in the recovery phase. This is characterized by vascular organization, restoration of BBB integrity, and by anti-inflammatory and reparative immune activities. Treatment with MSCs during the hyperacute and acute phases reduces cytotoxic damage by the production of neurotrophic factors, by the decrease in the pro-inflammatory mediators and immune infiltration and by the recovery of BBB integrity. During the subacute and chronic stages, MSC treatment may collaborate with the endogenous mechanisms of recovery by keeping immunomodulatory properties and by favoring vascular stabilization. CNS: central nervous system; ECM: extracellular matrix; BMVECs: brain microvascular endothelial cells; ECAMs: endothelial cell adhesion molecules; NK: natural killer; MMP: metalloproteinases; VEGF: vascular endothelial growth factor; TJ: tight junctions; Ang: angiopoietin; Tie2: tyrosine kinase receptor. Blue and red arrows mean increase and reduction, respectively.
Figure 5. Scheme illustrating pathophysiology of blood-brain barrier permeability during stroke. Left panel, hyperacute and acute stages are characterized by endothelial cell destabilization and BBB disruption, edema, peripheral immune infiltration, and glial immune activation. Right panel, subacute and chronic stages are involved in the recovery phase. This is characterized by vascular organization, restoration of BBB integrity, and by anti-inflammatory and reparative immune activities. Treatment with MSCs during the hyperacute and acute phases reduces cytotoxic damage by the production of neurotrophic factors, by the decrease in the pro-inflammatory mediators and immune infiltration and by the recovery of BBB integrity. During the subacute and chronic stages, MSC treatment may collaborate with the endogenous mechanisms of recovery by keeping immunomodulatory properties and by favoring vascular stabilization. CNS: central nervous system; ECM: extracellular matrix; BMVECs: brain microvascular endothelial cells; ECAMs: endothelial cell adhesion molecules; NK: natural killer; MMP: metalloproteinases; VEGF: vascular endothelial growth factor; TJ: tight junctions; Ang: angiopoietin; Tie2: tyrosine kinase receptor. Blue and red arrows mean increase and reduction, respectively.
Ijms 24 14117 g005
Table 2. Mesenchymal Stromal Cell-Studies selected from ClinicalTrials.gov for “Multiple Sclerosis”, “Ischemic Stroke”, “Alzheimer disease”, and “Parkinson disease”.
Table 2. Mesenchymal Stromal Cell-Studies selected from ClinicalTrials.gov for “Multiple Sclerosis”, “Ischemic Stroke”, “Alzheimer disease”, and “Parkinson disease”.
Components in the Clinical TrialsCategoriesStudies (%)
MSC typeBone Marrow27 (30)
Umbilical Cord24 (26.67)
Adipose14 (15.56)
Neural Progenitor-derived4 (4.44)
Embryonic1 (1.11)
Exosomes1 (1.11)
Not indicated20 (20.22)
Disorders/ConditionsMultiple Sclerosis35 (38.89)
Ischemic Stroke25 (27.78)
Alzheimer17 (18.89)
Parkinson13 (14.44)
ModalityAutologous41 (45.56)
Allogenic21 (23.33)
Not indicated28 (31.11)
RouteIntravenous48 (53.33)
Intrathecal8 (8.89)
Intravenous/Intrathecal3 (3.33)
Intraventricular1 (1.11)
Intra-striatal1 (1.11)
Intracerebral1 (1.11)
Nasal1 (1.11)
Not indicated27 (30)
TargetScore76 (76.77)
Immune13 (13.13)
Neurological10 (10.10)
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Vargas-Rodríguez, P.; Cuenca-Martagón, A.; Castillo-González, J.; Serrano-Martínez, I.; Luque, R.M.; Delgado, M.; González-Rey, E. Novel Therapeutic Opportunities for Neurodegenerative Diseases with Mesenchymal Stem Cells: The Focus on Modulating the Blood-Brain Barrier. Int. J. Mol. Sci. 2023, 24, 14117. https://doi.org/10.3390/ijms241814117

AMA Style

Vargas-Rodríguez P, Cuenca-Martagón A, Castillo-González J, Serrano-Martínez I, Luque RM, Delgado M, González-Rey E. Novel Therapeutic Opportunities for Neurodegenerative Diseases with Mesenchymal Stem Cells: The Focus on Modulating the Blood-Brain Barrier. International Journal of Molecular Sciences. 2023; 24(18):14117. https://doi.org/10.3390/ijms241814117

Chicago/Turabian Style

Vargas-Rodríguez, Pablo, Alejandro Cuenca-Martagón, Julia Castillo-González, Ignacio Serrano-Martínez, Raúl M. Luque, Mario Delgado, and Elena González-Rey. 2023. "Novel Therapeutic Opportunities for Neurodegenerative Diseases with Mesenchymal Stem Cells: The Focus on Modulating the Blood-Brain Barrier" International Journal of Molecular Sciences 24, no. 18: 14117. https://doi.org/10.3390/ijms241814117

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop