Next Article in Journal
Assessment of Coastal Winds in Iceland Using Sentinel-1, Reanalysis, and MET Observations
Previous Article in Journal
Effects of Preservation Methods on the Volatile Compound Profile and Physicochemical Properties of Aronia melanocarpa Berries
Previous Article in Special Issue
Effect of EVT-Derived Small Extracellular Vesicles on Normal and Impaired Human Implantation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Mesenchymal Stem Cells in Liver Fibrosis: A Dose-Dependent Recovery

1
Research and Education Resource Center for Cellular Technologies, Peoples Friendship University (RUDN University), 117198 Moscow, Russia
2
Shumakov National Medical Research Center of Transplantology and Artificial Organs, 125080 Moscow, Russia
3
Department of Human Anatomy and Histology, Department of Industrial Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
These authors also contributed equally to this work.
Appl. Sci. 2025, 15(19), 10471; https://doi.org/10.3390/app151910471 (registering DOI)
Submission received: 27 July 2025 / Revised: 20 September 2025 / Accepted: 23 September 2025 / Published: 27 September 2025
(This article belongs to the Special Issue Cell Biology: Latest Advances and Prospects)

Abstract

Mesenchymal stem cells (MSCs) are known to assist liver regeneration. In this study, we show a dose-dependent mode of recovery from liver fibrosis after intravenous injections of MSCs. Male Wistar rats experienced a 42-day-long modeling of liver fibrosis via CCl4 poisoning and received either a single injection of 2.5 × 106 MSCs on Day 3 after the last CCl4 dose or two MSC injections on Days 3 and 10. We dynamically monitored levels of liver cytolysis markers and cytokines in the venous blood and performed a histological study of Mallory-stained liver sections. All experimental groups experienced a nearly complete recovery of biochemical markers up to 4 weeks after the end of CCl4 administration, although we observed anti-inflammatory changes in the cytokine levels only in animals treated by two MSC injections. Histological study revealed minor signs of liver damage up to Day 90 in animals receiving two MSC doses with worse pathology in those who received a single MSC dose. Morphometric values stayed consistent with visual data, demonstrating a significantly larger number of binuclear hepatocytes, a smaller number of false lobules, and a lesser area of connective tissue proper in animals treated by two MSC injections. Our results reflect MSC grafting in applied doses to affect liver fibrosis in a dose-dependent mode. These findings provide a deeper understanding of MSC action in liver fibrosis, and the doses applied may serve as a milestone for further studies in humans.

1. Introduction

Liver fibrosis accompanies numerous cases of chronic liver disease, worsening the prognosis for survival. Epidemiologic studies show a broad prevalence of this pathology. For instance, advanced liver fibrosis accounts for 2.85% of adults in mainland China [1]. Despite the obvious severity of replacing normal liver tissue with connective tissue proper, liver fibrosis may regress, although this process is slow and challenging [2,3,4].
During the past ten years, a growing body of evidence has disclosed more components of the machinery for such a recovery from liver fibrosis. Briefly, both metabolic regulation and cellular immunity play important roles, especially in stimulation by bone marrow stem cells, although via different mechanisms for hematopoietic stem cells (HSCs) and mesenchymal stem cells (MSCs). For the hematopoietic factor, molecular pathways of regulation involve tumor necrosis factor (TNF)-related, apoptosis-induced ligand binding in apoptotic changes in HSCs, and natural killers affect the HSC effects via gamma interferon (IFN-γ) action, whereas HSC-derived macrophages govern the production of matrix metalloproteinases to prevent the excessive proliferation of fibroblasts [5].
MSCs are also well known for preventing further fibrosis development [6,7,8,9,10,11] and even enhancing the formation of new hepatocyte-like cells via various machinery involved [12]. MSCs affect the proliferation of perisinusoidal stellate cells, transforming them into myofibroblasts and reducing collagen production [13]. Furthermore, MSCs, located in the liver, regulate neighboring cells in a paracrine way by releasing exosomes [14]. Recently, it has been shown in studies of a murine model that MSC-derived exosomes reduce endoplasmic reticulum stress and increase autophagic flux [15].
Due to this multifaceted activity of MSCs in the liver, their grafting is a prominent frontier for medical research [16]. However, few clinical studies are accessible for MSCs in liver fibrosis, except for liver cirrhosis. In cirrhotic patients, eleven clinical studies have already been accomplished, revealing a beneficial impact on survival and quality of life [17]. Nevertheless, these reports do not focus on other reasons for liver fibrosis, especially relevant for non-alcohol fatty liver disease or autoimmune pathology, implying possible graft-versus-host reactions in MSC grafting.
To design clinical approaches in the future, there exists a need for a more comprehensive understanding of common animal models (e.g., rats) of MSC usage. In this study, we aimed to study which time- and dose-dependent biochemical and histological changes occur in liver fibrosis after MSC grafting at early terms and whether there generally exists a considerable dose dependency (Figure 1). For this, we focused on bone marrow-derived MSCs as the most appropriate for potential clinical extrapolation rather than amniotic or umbilical cord-derived stem cells and hypothesized over whether there is a bond between the dose regimen of bone marrow MSCs and the mode of recovery from liver fibrosis.

2. Materials and Methods

2.1. Animals

This experimental study involved 96 male Wistar rats of 6–8 months of age with body masses of 400–450 g. The rodents stayed in our vivarium at 18–22 °C with a mixed diet and free access to water. The experimental procedures with animals took place in daytime (9 to 12 am) to avoid fluctuations in mitotic activity in the liver. All the manipulations were carried out in accordance with international and local rules (European Convention for the Protection of Vertebrate Animals Used for Experimental and Other Scientific Purposes, Russian State Standards GOST 33215-2014 and GOST 33216-2014 [18,19]) and were approved by the local ethical committee of Federal State Budgetary Institution “Shumakov National Medical Research Center of Transplantology and Artificial Organs” of the Ministry of Health of the Russian Federation (protocol No. 050221-1/3e 05/02/21) for studies involving animals. Only healthy rats, after a 2-week-long quarantine and veterinarian check-up, were included in this study (a priori criteria); no exclusions were made after the study group was formed.

2.2. Mesenchymal Stem Cells

We obtained allogeneic MSCs from 24 intact male Wistar rat donors aged 6–8 months with body masses of 400–450 g. We avoided using MSCs from experimental rats due to their diminished adhesive and proliferative activity with excessive debris in primary cell cultures. After bone marrow aspiration, we centrifugated the samples to obtain stem cells and removed the remaining components. We cultivated the resulting cell fraction in Dulbecco’s modified Eagle medium with 10% fetal bovine serum, L-glutamine at 2 mmol/L, penicillin at 100 U/mL, and streptomycin at 100 U/mL. The cell cultivation proceeded at 37 °C in air containing 5% CO2 up to a high-confluent state (90%). Two passages of MSC cultivation were provided with overall cultivation term of 15 days. To check the phenotype of the cells, we provided a flow cytometric study of key superficial positive and negative markers (CD14, CD19, CD34, CD45, and HLA-DR should be negative for MSCs, whereas CD73, CD90, and CD105 should be positive) [20]. The proliferative potential of the MSCs was further improved by differentiation for various lineages of MSCs (StemPro Adipogenesis/Osteogenesis Differentiation Kits; Gibco; Jenks, OK, USA), with only samples of >95% trypan blue-positive cells being considered viable. Then, the cells were washed twice in isotonic saline and grafted onto the tail veins of the rats with insulin syringes.

2.3. Liver Fibrosis Modeling

We modeled liver fibrosis by poisoning Wistar rats (n = 72, not including the donor animals) for 42 days using carbon tetrachloride (CCl4). Briefly, we provided subcutaneous injections of 60% CCl4 and peach oil solution twice a week (Monday and Thursday) at a dose of 0.3 mL per 100 g of animal body mass, although the first injection contained 0.5 mL of 60% CCl4 per 100 g of animal body mass. Therefore, the cumulative dose of CCl4 stayed equal to 3.5 mL per 100 g of animal body mass.

2.4. Study Groups

After the modeling of fibrosis, we randomized the rats (n = 72) into experimental groups by the method of random numbers. Animals of Group 1 (n = 24) received a single intravenous injection of 2.5 × 106 MSCs in 1 mL of saline on Day 3 after the end of CCl4 treatment. Dosage regimens were regarded with respect to doses tested in our pilot experiments, revealing a sharp increase in efficacy at the range of 2.5–5 × 106 MSCs per animal. In Group 2 (n = 24), rats were injected twice with the same dose of MSCs on Days 3 and 10 after the last CCL4 treatment (both doses of MSCs were obtained from the same donor in all cases). Group 3 (control, n = 24) received no MSCs but an intravenous injection of 1 mL saline, i.e., sham treatment. The endpoints for blood sampling were on Days 14, 21, and 28 (plus Day 35 for cytokines), whereas the histological endpoints were on Days 3, 7, 14, 21, 28, 60 (three animals from group per point), and 90 (the rest). We sacrificed the animals via perfusion fixation by neutral buffered formalin and anesthesia by Zoletil-100 (Virbac, Carros, France) followed by liver autopsy with further histological analysis.

2.5. Blood Tests

Every seven days, we performed blood tests to follow the dynamics of biochemical markers for cytolysis in the liver, as well as for pro- and anti-inflammatory cytokines. To obtain blood from tail veins, the rats were anesthetized, and their tail tips were clipped. In the peripheral blood, we measured levels of biochemical liver markers: alanine aminotransferase (ALT), aspartate aminotransferase (AST), and alkaline phosphatase (ALP). The quantitative analysis was performed using a Reflotron biochemical analyzer and test strips from the same manufacturer (Reflotron; Roche, Switzerland; AST–107 45 120, ALT–107 45 138, ALP–116 22 773, respectively). Furthermore, we studied the blood levels of interleukins, pro-inflammatory IL-4 and anti-inflammatory IL-10, as well as such pro-inflammatory cytokines such as IFN-γ and TNF-α by enzyme-linked immunosorbent assay (ELISA) with appropriate test systems (Protein contour; Saint-Petersburg, Russia; Vector-Best; Novosibirsk, Russia) using a ChemWell analyzer (Awareness Technology; Palm City, FL, USA).

2.6. Histological Study

Immediately after the rats were euthanized, we fixed the samples of liver tissue in 10% neutral buffered formalin and, after a standard dehydration, embedded them into paraffin. We provided Mallory’s trichrome staining at sections of 5 µm for histological study with easy detection of collagen fibers. For these sections, we measured the degree of fibrosis with ImageScopeM 12.1 software (systems for microscopy and analysis; Moscow, Russia) using microphotographs obtained with a Leica DM1000 microscope and a Leica LTDCH9435 DFC 295 camera (Leica; Wetzlar, Germany). For this, we calculated the percentage of blue area on Mallory-stained slides per field of view at microscope magnification ×400. Also, we counted the number of false lobules and binuclear hepatocytes per field of view of the same size.
For immunofluorescent study, we employed anti-Ki-67 antibodies (clone SR00-02, lot No. HA721115, article HP0304; Huabio, PRC, Woburn, MA, USA) at 1:100 PBS dilution. After incubation, we washed the sections with PBS and then processed them with secondary polyclonal antibodies, conjugated with horseradish peroxidase (Anti-Rabbit-HRP, lot No. HA1119, article M05-22-P2; Huabio, PRC). DAB served as chromogen. Data analysis was performed with blinded data by contributors who did not work directly with the experimental animals.

2.7. Statistical Analysis

Sample size was calculated with respect to pilot experiments as the least number of animals to obtain statistical significance in morphometrical values with a power of more than 0.8. We processed the resulting numeric data with OriginPro 2024 software (OriginLab; Northampton, MA, USA) by distribution normality assessment with Shapiro–Wilk test and following one-factor analysis of variance (ANOVA) together with Bonferroni correction, as all the numeric data samples followed normal distribution. The differences between the group samples were considered significant for p < 0.05 and a statistical power of more than 0.8.

3. Results

3.1. MSCs Are the Principal Component of Cell Culture in This Study

We cultivated cells of the MSC line obtained from the bone marrow of healthy rat donors. We validated the lineage of MSCs by flow cytometry. First, we increased the signal-to-noise ratio by excluding debris and doublets (Figure 2A). We picked data for including cells into calculations and found out that cell culture corresponds to the aforementioned criteria of MSC phenotype (Figure 2B). Furthermore, we provided an increase in proliferative potential through differentiation stimulation for osteogenic and adipogenic lineages (Figure 3). Then, we modeled the liver fibrosis.

3.2. Biochemical Markers of Liver Cytolysis Recover Faster After MSC Grafting

Immediately after the last day of CCl4 poisoning, all surviving rats had a sharp increase in blood markers for liver damage, i.e., ALT, AST, and ALP (Figure 4A). Further dynamics of levels were similar for these three markers but rather different for the study groups. The animals from Group 1 received a single dose of MSCs on Day 3, and Group 2, which experienced double intravenous grafting of MSCs on Days 3 and 10, showed a significant decline in ALP and ALT levels up to Day 14, almost to initial values. These levels stayed approximately the same until the end of the blood test timeline with means for Group 2 being slightly less than in Group 1, although not significantly. Control rats from Group 3 also tended to recover their levels of biochemical markers. However, this recovery was less remarkable than in Groups 1 and 2, and for AST no full recovery occurred.

3.3. Cytokine Levels Vary Greatly After MSC Grafting

The largest number of inconsistencies between the groups and timepoints were detected for dynamics of cytokine levels, especially pro-inflammatory ones (Figure 4B). Thus, IL-10, an anti-inflammatory cytokine, revealed an exponential growth in animals’ blood throughout the time of the study, although the peak of IL-10 by Day 35 was significantly higher in Group 2. Interestingly, almost the same pattern of time-dependent changes is typical for IL-4 levels, which is basically a pro-inflammatory cytokine.
Meanwhile, Group 2 is remarkable for a specific mode of level changes for IFN-γ and TNF-α. The content of both these markers in control animals tended to increase up to Day 21 and then declined nearly to the initial levels. In contrast, Groups 1 and 3 (MSC-treated with a single dose and not treated, respectively) had a slighter rise in IFN-γ and TNF-α. Nevertheless, the content of these markers for Groups 1 and 2 held an uptrend until the last test, with the graph passing significantly upwards from Group 3, as seen in Figure 4B.

3.4. MSC Grafting Ameliorates Histological Signs of Liver Damage

On the histological slides, stained by Mallory’s trichrome, animals of all groups demonstrated considerably impaired histoarchitectonic traits in the liver up to Day 14 after the end of liver fibrosis modeling (Figure 5A, the line above; arrows mark the changes mentioned further). Specifically, the microphotographs for Day 14 demonstrate that the structure of hepatic plates is poorly seen. Furthermore, there is evident blood congestion in the sinusoidal capillaries as they widen, which is more remarkable in the pericentral zones of hepatic lobules. False lobules and numerous adipocytes are also found. Collagen fibers of connective tissue proper, replacing normal liver tissue, are visualized well due to their blueish color. No infiltration by inflammatory cells is seen.
However, the morphological pattern changes differently in study groups up to three months after the grafting. The slide microphotograph for a rat from Group 1 (Figure 5A, the line below), which received a single MSC injection, reveals a notable improvement in liver histology. Thus, no adipocytes are found, and hepatic plates are visible, although strong blood congestion still exists when central veins and sinusoidal capillaries are enlarged. Group 2, treated by double MSC doses, has a better image of histological changes with almost complete recovery of the aforementioned quantitative parameters. In Group 3, including animals after the sham treatment with saline, morphological features on Day 90 do not differ greatly from those on Day 14.
In the histological study, the visual differences between the groups are consistent with calculated morphometric values. The number of false lobules (Figure 5B) increases in all animals with liver fibrosis, with nearly complete recovery to sporadic false lobules found in Group 2 (two MSC injections) and a slight decrease in Group 1 (single MSC injection), which is significantly different from Group 3 (sham treatment after CCl4) with practically no recovery at all. The percentage of connective tissue area followed the same tendency (Figure 5C), except for the sharp turn downwards in Group 2. Therefore, at 90 days since fibrosis modeling, Group 2 experienced full recovery with excessive connective tissue disappearing, whereas Group 1 had partial recovery, and Group 3 had no recovery at all. The last morphometric parameter measured, the number of binuclear hepatocytes, followed the pattern of a gradual, exponential increase for all the groups (Figure 5D), although more pronouncedly for Group 2 after double MSC injection.
Furthermore, we have provided immunohistochemical/immunofluorescent study to observe whether MSC-treated groups have another rate of expression of markers for regeneration. For this, we employed anti-Ki67 immunofluorescent staining. Ki67-positive cells already appeared in Group 1 after being treated by a lesser amount of MSCs (Figure 6A), whereas their number increases in Group 2, treated by a larger dose of MSCs. Meanwhile, animals with fibrosis but no further treatment (Group 3), as well as healthy donors, experience almost no Ki67 expression at all. For the statistical calculations (Figure 6B), Groups 1 and 2 have a significantly larger number of Ki67-positive cells per field of view (p < 0.05) than Group 3. Therefore, the IHC-related data are consistent with the histochemical ones.

4. Discussion

In our study, we observed a dose-dependent recovery from liver fibrosis through histological and biochemical data after 6 weeks of CCl4 poisoning. The dynamics of ALT, ALP, and AST revealed no unexpected findings, though they approved of the model and showed dose-dependent differences. The only point we should outline is the relatively rapid recovery of liver cytolysis markers even in the case of no actual treatment, which demonstrates the liver’s fascinating potential regeneration. Meanwhile, another cluster of blood testing data, the cytokine study, has provided us with more interesting outputs.
Animals from Group 2, receiving a double dose of MSC grafting, were remarkable for an initial increase in IFN-γ and TNF-α levels up to Day 21 and then a reversal almost to the baseline, a feature not observed in Groups 1 and 3 (one dose of MSCs and sham MSCs, correspondingly) with a slight but constant increase in IFN-γ and TNF-α content until the end of the experimental study. This pattern of IFN-γ and TNF-α is perhaps related to the recruitment of HSC-associated molecular pathways and the direct involvement of regulation by natural killers [5]. However, the role of HSCs should not be overestimated here. We consider that there exists a probable interaction between MSCs and T cells via exosomes in our experiments [21]. Furthermore, not only exosomes may mediate the effects but mitochondria too, if they are transported from MSCs into T cells [22]. This codependence between stem cells and T cells requires an elucidation in future studies by additional methods.
For interleukins, we observed two similar trends in both pro-inflammatory IL-4 and anti-inflammatory IL-10, although significantly less pronounced in MSC-treated Groups 1 and 2. While the dynamics of IL-4 are consistent with well-known data, the sharp peak of IL-10 up to Day 35 in animals with sham treatment (Group 3) demands discussion. We assume this CD10 peak is compensatory and beneficial but simply not required in the presence of MSC stimulation. In animals without treatment, CD10-related machinery may induce immune modulation via substance P-associated regulation [23], thereby supporting weak positive dynamics. Concerning the immune effects, we should also emphasize that all the changes seen were only biochemical, and no infiltration by leukocytes has been revealed on histological slides.
However, histological sections of the liver visualize the difference between single and double doses of MSCs (Figure 5), which is consistent with morphometric data from both Mallory’s and anti-Ki67 staining. Calculations showed that the fibrosis degree decreases gradually with a significant dose dependence, as seen from the reduced number of false lobules in MSC-treated groups and the diminished percentage of connective tissue proper found there. We also observed a larger number of binuclear hepatocytes and Ki67+ cells. Importantly, binuclear hepatocytes increase at many timepoints for Group 3 (not treated by MSCs) and even more than in Group 2 (treated by two doses). We believe this is due to a more rapid recovery of fibrosis in Group 2, requiring less healthy hepatocytes to experience a polyploid transformation. However, we avoided considering the increase in binucleated hepatocytes as an event of proliferation of hepatocytes, as it may just be a compensatory hypertrophy of the remaining functional portion of the liver. Indeed, we have not observed mitosis figures nor angiogenesis that could be related to de novo hyperplastic changes.
Furthermore, the remarkable regeneration with a normal histoarchitectonic structure formed de novo in double MSC grafting should be due not only to regressing fibrosis, survival of hepatocytes, or forming new hepatocyte-like cells but also to a recovery of hepatic plates and sinusoidal capillaries between them. It has been reported that MSCs may transform into endothelial cells [24] in vivo in cardiovascular pathology with a beneficial impact on its course. Thus, one may expect the same effect in enhancing the repair of hepatic plates, and that requires additional investigation in the future. Moreover, we may anticipate a direct link between the cytokine and histological changes observed, especially in the reconstruction of hepatic plates and sinusoidal capillaries.
Generally, our study showed MSCs’ well-known potential for liver regeneration but with novel data on the rigorous dose-dependent mode of such an impact observed both histochemically and immunohistochemically. Moreover, we observed that systemic administration is also beneficial for MSC activity in the liver, despite previously reported facts, although in mice and not in rats [8]. A similar dose-dependent mode of MSCs was revealed earlier in other pathologies, such as in acute kidney injury [25].
In this study, we intended to regard the impact of MSCs without any applicable support, like cell-engineered constructs involved to maintain their survival. Nevertheless, the dose-dependent effects of MSC usage in applied doses are well found. However, we do not assume a direct impact of MSCs on fibrosis in the liver. We consider the observed changes to be due to paracrine, exosomal, and immune-modulating regulation, as shown previously [26].
Generally, exosomal influence of grafted MSCs is considered the most prominent, with many RNAs transferred. For example, microRNAs such as miR-21-3p enhance endothelial growth and suppress fibroblastic proliferation through inhibition of phosphatase and tensin homologs, as well as sprouty homolog 1 (reported for umbilical MSCs) [27], while several RNAs act via the PI3K-AKT-mTOR axis (reported for amniotic MSCs) [28]. Moreover, both molecular machineries affect macrophage activity. Further, bone marrow-derived MSCs, relevant for our study, were also shown to include tumor growth factor beta and SMAD-protein-related mechanisms, thus preventing excessive connective tissue formation and favoring regeneration, and this is in skin wound healing [29]. Nevertheless, exosomal release alone is probably not sufficient to provide all the beneficial effects of bone marrow MSCs: in a model of renoprotection, bone marrow MSCs had a better impact on fibrosis than their separated exosomes [30].
Specially for liver fibrosis models of bone marrow MSC grafting, some supplementary machinery has been reported. First of all, not exosomes only but microvesicles of MSCs permit the release of active factors from MSCs [31]. Thus, it has been recently shown that the regenerative potential of bone marrow MSCs in the liver depends on protein tyrosine phosphatase type 4A activity. If it is strong in the mitochondria of MSCs, they can benefit from anaerobic metabolism [32]. Interestingly, the efficiency of bone marrow MSC grafting in the liver also correlates with erythropoietin levels [33]. Furthermore, matrix metalloproteinase 14 can serve as a marker of successful bone marrow MSC action in liver regeneration, as its levels increase due to excessive secretion by surviving MSCs [34]. Then, immune mechanisms are involved in the bone marrow MSC-associated effects in the liver. Indeed, NK cells increase in the liver after bone marrow MSC grafting, accelerating the removal of impaired cells [35].
Furthermore, research with stem cells raises an important issue of ensuring their viability and efficiency for successful grafting. For example, a lot of previous research, including ours, has reported possible approaches to improving the viability of MSCs via artificially designed gels [36,37,38]. However, the practical choice between MSCs and HSCs may be challenging in possible clinical implementation, as we showed the benefit of HSCs and their total RNA especially in chronic liver disease [39]. We consider this choice should be based on potential adverse effects in certain pathology and not on the direct impact of MSCs or HSCs.

Limitations and Perspectives

  • The set of diagnostic tools involved is limited. For instance, a recent study with a similar design, although via 9 and not 6 weeks of CCl4 poisoning, revealed antioxidant machinery involved in the MSC effects in liver fibrosis [40]. This research, parallel to ours, implies the need to enlarge the set of methods used, as the current study was obviously limited and may be even biased by the use of fixed tissues not able to reveal the molecular pathways of antioxidation. We propose that an aspiration biopsy via a known method [41] can be appropriate for the widening of research in this field. Hypoxic effects also deserve a detailed study because of their ambiguity in liver fibrosis. For example, a fascinating report by Kuo and colleagues showed MSCs from Wharton’s jelly were able to reverse fibrotic changes in the liver, and this impact has been more evident in hypoxic states [42]. Furthermore, future research should include additional methods of liver fibrosis visualization with higher resolution and/or accuracy (e.g., coherent anti-Stokes Raman scattering microscopy, fluorescence lifetime imaging, or second harmonic generation microscopy) to observe the dynamic recovery of hepatic plates.
  • Our study does not consider the variety of subpopulations for MSCs [43]. Perhaps, a more refined selection of MSC subtypes may lead to a more remarkable beneficial impact. For example, Nishina and colleagues have recently demonstrated that Muse cells, a subpopulation of MSCs, exhibit the largest proliferative potential in liver fibrosis after twelve weeks of CCl4 poisoning [44]. Furthermore, not only are MSC subpopulations not regarded in the current work but also immune- and apoptosis-related markers, as the study design was focused on revealing whether there exists a dose-dependent effect. However, further research should elucidate the issue of certain apoptotic and immune-mediated pathways involved in the liver’s recovery from fibrosis in this model.
  • The choice of intervention mode for fibrosis modeling is challenging. We preferred CCl4, as it is the most common agent for fibrosis modeling in animals [45], not only with rats but mice [46] and monkeys [47], too. Furthermore, we had tested it successfully in our previous studies [37,38]. However, other chemical models than CCl4 have also been successfully tested in rats, for example, poisoning with thioacetamide [48], diethylnitrosamine, and acetaminophen [45]. Furthermore, surgical, organelle-targeting, immune-mediated, alcohol- or diet-provoked models exist, as well as liver fibrosis modeling via breeding transgenic animals with related genes involved [45]. All these modes require additional testing of our regimen for liver fibrosis recovery in our further research.
  • Animals of the same sex were included to obtain consistent results; for ethical reasons, we avoided doubling animal number with both sexes observed. Thus, we preferred male rats, as comparable experiments with male rats have been conducted by other teams [49,50,51]. However, we assume female rats to be also appropriate for such studies, and further research should also account for testing MSCs in female animals.
  • There exists a necessity to entirely and comprehensively describe if the MSC-induced changes permit survival of hepatocytes with their compensatory hypertrophy only or if certain hyperplasia can also be seen. In further studies, a detailed evaluation of cell growth is therefore required.
  • An additional limitation raises the possibility of extrapolation between our studies and human disease. For sure, conventional models of liver fibrosis have already been regarded as partially available for translation to human pathology [52]. To bring these models closer to human liver structure, liver fibrosis models have been developed in primates in recent years [47,53]. However, not only do the dosage regimens between rodent/primate studies and clinical studies of MSCs in liver pathology differ, but also it is challenging in clinical studies to find a proper dosage (various clinical studies used doses differing in billions of ways) [54,55,56]; thus, the importance of our study is of high level.

5. Conclusions

In this study, we aimed to reveal if MSC grafting has an impact on liver fibrosis in a rat model at early terms. We employed easy-to-access biochemical and histological methods and therefore demonstrated a successful model of liver damage and further recovery consistent with parallel studies in the field. Importantly, we revealed a remarkable dose-dependent impact of MSC grafting on the course of liver fibrosis as well as on accompanying levels of liver enzymes and cytokines. Further studies with respect to the perspectives and limitations mentioned are required for a comprehensive understanding of fascinating MSC potential.

Author Contributions

A.L.: conceptualization, validation, formal analysis, data curation, and writing—original draft preparation; M.S.: conceptualization, validation, formal analysis, data curation, writing—original draft preparation, and writing—review and editing; N.O.: conceptualization, methodology, validation, formal analysis, data curation, writing—original draft preparation, and supervision; V.B.: conceptualization and supervision; M.K.: validation, formal analysis, and data curation; A.V.: data curation, writing—original draft preparation, and writing—review and editing; K.P.: data curation and writing—review and editing; A.N.: methodology and data curation; E.K.: formal analysis and data curation; A.K.: data curation, supervision, and funding acquisition; A.A.: validation and writing—review and editing; P.F.: validation and writing—review and editing; N.K.: formal analysis and data curation; G.P.: formal analysis, data curation, writing—original draft preparation, writing—review and editing, supervision, and project administration; S.G.: formal analysis, data curation, writing—original draft preparation, writing—review and editing, supervision, and project administration. All authors have read and agreed to the published version of the manuscript.

Funding

This study was performed with a support of the state assignment of the Ministry of Science and Higher Education of the Russian Federation (Grant No. 075-15-2022-310).

Institutional Review Board Statement

The animal study protocol was approved by the Ethics Committee of Federal State Budgetary Institution “Shumakov National Medical Research Center of Transplantology and Artificial Organs” of the Ministry of Health of the Russian Federation (protocol 050221-1/3e 05/02/21) for studies involving animals.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding authors.

Acknowledgments

The authors are grateful to Deev R. and Gazizov I. for research assistance.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Man, S.; Deng, Y.; Ma, Y.; Fu, J.; Bao, H.; Yu, C.; Lv, J.; Liu, H.; Wang, B.; Li, L. Prevalence of liver steatosis and fibrosis in the general population and various high-risk populations: A nationwide study with 5.7 million adults in China. Gastroenterology 2023, 165, 1025–1040. [Google Scholar] [CrossRef] [PubMed]
  2. Terai, S.; Tsuchiya, A. Status of and candidates for cell therapy in liver cirrhosis: Overcoming the “point of no return” in advanced liver cirrhosis. J. Gastroenterol. 2017, 52, 129–140. [Google Scholar] [CrossRef] [PubMed]
  3. Epstein, S.E.; Luger, D.; Lipinski, M.J. Large animal model efficacy testing is needed prior to launch of a stem cell clinical trial: An evidence-lacking conclusion based on conjecture. Circ. Res. 2017, 121, 496–498. [Google Scholar] [CrossRef]
  4. Messina, A.; Luce, E.; Hussein, M.; Dubart-Kupperschmitt, A. Pluripotent-stem-cell-derived hepatic cells: Hepatocytes and organoids for liver therapy and regeneration. Cells 2020, 9, 420. [Google Scholar] [CrossRef]
  5. Roehlen, N.; Crouchet, E.; Baumert, T.F. Liver fibrosis: Mechanistic concepts and therapeutic perspectives. Cells 2020, 9, 875. [Google Scholar] [CrossRef]
  6. Liu, P.; Mao, Y.; Xie, Y.; Wei, J.; Yao, J. Stem cells for treatment of liver fibrosis/cirrhosis: Clinical progress and therapeutic potential. Stem Cell Res. Ther. 2022, 13, 356. [Google Scholar] [CrossRef] [PubMed]
  7. Zhao, L.; Chen, S.; Shi, X.; Cao, H.; Li, L. A pooled analysis of mesenchymal stem cell-based therapy for liver disease. Stem Cell Res. Ther. 2018, 9, 72. [Google Scholar] [CrossRef]
  8. Van der Helm, D.; Barnhoorn, M.C.; de Jonge-Muller, E.S.M.; Molendijk, I.; Hawinkels, L.J.A.C.; Coenraad, M.J.; van Hoek, B.; Verspaget, H.W. Local but not systemic administration of mesenchymal stromal cells ameliorates fibrogenesis in regenerating livers. J. Cell Mol. Med. 2019, 23, 6238–6250. [Google Scholar] [CrossRef]
  9. Zhou, G.P.; Jiang, Y.Z.; Sun, L.Y.; Zhu, Z.J. Therapeutic effect and safety of stem cell therapy for chronic liver disease: A systematic review and meta-analysis of randomized controlled trials. Stem Cell Res. Ther. 2020, 11, 419. [Google Scholar] [CrossRef]
  10. Zhang, Y.; Li, Y.; Zhang, L.; Li, J.; Zhu, Z. Mesenchymal stem cells: Potential application for the treatment of hepatic cirrhosis. Stem Cell Res. Ther. 2018, 9, 59. [Google Scholar] [CrossRef]
  11. Shi, M.; Li, Y.Y.; Xu, R.N.; Meng, F.P.; Yu, S.J.; Fu, J.L.; Hu, J.H.; Li, J.X.; Wang, L.F.; Jin, L.; et al. Mesenchymal stem cell therapy in decompensated liver cirrhosis: A long-term follow-up analysis of the randomized controlled clinical trial. Hepatol. Int. 2021, 15, 1431–1441. [Google Scholar] [CrossRef]
  12. Yang, S.; Liu, J.; Khe, J.S.; Lu, A.J.H.; Lim, V. Emerging insights into mesenchymal stem cells and exosome-based therapies for liver injury. Biomol. Biomed. 2025, 25, 1691–1708. [Google Scholar] [CrossRef]
  13. Hisada, M.; Zhang, X.; Ota, Y.; Cameron, A.M.; Burdick, J.; Gao, B.; Williams, G.M.; Sun, Z. Fibrosis in small syngeneic rat liver grafts because of damaged bone marrow stem cells from chronic alcohol consumption. Liver Transpl. 2017, 23, 1564–1576. [Google Scholar] [CrossRef] [PubMed]
  14. Wang, F.D.; Chen, E.Q. Mesenchymal stem cell-derived exosomes in the treatment of end-stage liver disease. Curr. Stem Cell Res. Ther. 2025, in press. [Google Scholar] [CrossRef] [PubMed]
  15. Moayedfard, Z.; Bagheri, L.K.; Alizadeh, A.A.; Nekooeian, A.A.; Dara, M.; Koohpeyma, F.; Parsa, S.; Nikeghbalian, S.; Hosseinpouri, A.; Azarpira, N. The ameliorative effect of adipose-derived mesenchymal stem cells and their exosomes in non-alcoholic steatohepatitis by simultaneously enhancing autophagic flux and suppressing endoplasmic reticulum stress. Iran. J. Med. Sci. 2025, 50, 334–350. [Google Scholar] [CrossRef] [PubMed]
  16. Ruiz, M.A.; Kaiser Junior, R.L.R.; Piron-Ruiz, G.; de Quadros, L.G. Are mesenchymal stem/stromal cells a novel avenue for the treatment of non-alcoholic fatty liver disease? World J. Stem Cells. 2025, 17, 99638. [Google Scholar] [CrossRef]
  17. Pînzariu, A.C.; Moscalu, R.; Soroceanu, R.P.; Maranduca, M.A.; Drochioi, I.C.; Vlasceanu, V.I.; Timofeiov, S.; Timofte, D.V.; Huzum, B.; Moscalu, M.; et al. The therapeutic use and potential of MSCs: Advances in regenerative medicine. Int. J. Mol. Sci. 2025, 26, 3084. [Google Scholar] [CrossRef]
  18. GOST 33216-2014; Guidelines for Accommodation and Care of Animals. Species-Specific Provisions for Laboratory Rodents and Rabbits. Standartinform: Moscow, Russia, 2016.
  19. GOST 33215-2014; Guidelines for Accommodation and Care of Animals. Environment, Housing and Management. Standartinform: Moscow, Russia, 2019.
  20. Kuçi, Z.; Piede, N.; Vogelsang, K.; Pfeffermann, L.M.; Wehner, S.; Salzmann-Manrique, E.; Stais, M.; Kreyenberg, H.; Bonig, H.; Bader, P.; et al. Expression of HLA-DR by mesenchymal stromal cells in the platelet lysate era: An obsolete release criterion for MSCs? J. Transl. Med. 2024, 22, 39. [Google Scholar] [CrossRef]
  21. Fujii, S.; Miura, Y. Immunomodulatory and regenerative effects of MSC-derived extracellular vesicles to treat acute GVHD. Stem Cells 2022, 40, 977–990. [Google Scholar] [CrossRef]
  22. Court, A.C.; Le-Gatt, A.; Luz-Crawford, P.; Parra, E.; Aliaga-Tobar, V.; Bátiz, L.F.; Contreras, R.A.; Ortúzar, M.I.; Kurte, M.; Elizondo-Vega, R.; et al. Mitochondrial transfer from MSCs to T cells induces Treg differentiation and restricts inflammatory response. EMBO Rep. 2020, 21, e48052. [Google Scholar] [CrossRef]
  23. Kouroupis, D.; Bowles, A.C.; Willman, M.A.; Perucca Orfei, C.; Colombini, A.; Best, T.M.; Kaplan, L.D.; Correa, D. Infrapatellar fat pad-derived MSC response to inflammation and fibrosis induces an immunomodulatory phenotype involving CD10-mediated Substance P degradation. Sci. Rep. 2019, 9, 10864. [Google Scholar] [CrossRef] [PubMed]
  24. Ye, Z.Q.; Meng, X.H.; Fang, X.; Liu, H.Y.; Mwindadi, H.H. MiR-126 regulates the effect of mesenchymal stem cell vascular repair on carotid atherosclerosis through MAPK/ERK signaling pathway. World J. Stem Cells. 2025, 17, 106520. [Google Scholar] [CrossRef]
  25. Cao, J.Y.; Wang, B.; Tang, T.T.; Wen, Y.; Li, Z.L.; Feng, S.T.; Wu, M.; Liu, D.; Yin, D.; Ma, K.L.; et al. Exosomal miR-125b-5p deriving from mesenchymal stem cells promotes tubular repair by suppression of p53 in ischemic acute kidney injury. Theranostics 2021, 11, 5248–5266. [Google Scholar] [CrossRef]
  26. Yao, L.; Hu, X.; Dai, K.; Yuan, M.; Liu, P.; Zhang, Q.; Jiang, Y. Mesenchymal stromal cells: Promising treatment for liver cirrhosis. Stem Cell Res. Ther. 2022, 13, 308. [Google Scholar] [CrossRef]
  27. Hu, Y.; Rao, S.S.; Wang, Z.X.; Hu, Y.; Rao, S.S.; Wang, Z.X. Exosomes from human umbilical cord blood accelerate cutaneous wound healing through miR-21-3p-mediated promotion of angiogenesis and fibroblast function. Theranostics 2018, 8, 169–184. [Google Scholar] [CrossRef]
  28. Wei, P.; Zhong, C.; Yang, X.; Shu, F.; Xiao, S.; Gong, T.; Luo, P.; Li, L.; Chen, Z.; Zheng, Y.; et al. Exosomes derived from human amniotic epithelial cells accelerate diabetic wound healing via PI3K-AKT-mTOR-mediated promotion in angiogenesis and fibroblast function. Burns Trauma 2020, 8, tkaa020. [Google Scholar] [CrossRef]
  29. Jiang, T.; Wang, Z.; Sun, J. Human bone marrow mesenchymal stem cell-derived exosomes stimulate cutaneous wound healing mediates through TGF-β/Smad signaling pathway. Stem Cell Res. Ther. 2020, 11, 198. [Google Scholar] [CrossRef] [PubMed]
  30. Li, Y.; Chakraborty, A.; Broughton, B.R.S.; Ferens, D.; Widdop, R.E.; Ricardo, S.D.; Samuel, C.S. Comparing the renoprotective effects of BM-MSCs versus BM-MSC-exosomes, when combined with an anti-fibrotic drug, in hypertensive mice. Biomed. Pharmacother. 2021, 144, 112256. [Google Scholar] [CrossRef]
  31. Sabry, D.; Mohamed, A.; Monir, M.; Ibrahim, H.A. The Effect of Mesenchymal Stem Cells Derived Microvesicles on the Treatment of Experimental CCL4 Induced Liver Fibrosis in Rats. Int. J. Stem Cells. 2019, 12, 400–409. [Google Scholar] [CrossRef] [PubMed]
  32. Kim, J.Y.; Kim, S.H.; Seok, J.; Bae, S.H.; Hwang, S.G.; Kim, G.J. Increased PRL-1 in BM-derived MSCs triggers anaerobic metabolism via mitochondria in a cholestatic rat model. Mol. Ther. Nucleic Acid. 2023, 31, 512–524. [Google Scholar] [CrossRef]
  33. Wang, X.; Wang, H.; Lu, J.; Feng, Z.; Liu, Z.; Song, H.; Wang, H.; Zhou, Y.; Xu, J. Erythropoietin-Modified Mesenchymal Stem Cells Enhance Anti-Fibrosis Efficacy in Mouse Liver Fibrosis Model. Tissue Eng. Regen. Med. 2020, 17, 683–693. [Google Scholar] [CrossRef]
  34. Fukushima, K.; Itaba, N.; Kono, Y.; Fukushima, K.; Itaba, N.; Kono, Y.; Okazaki, S.; Enokida, S.; Kuranobu, N.; Murakami, J.; et al. Secreted matrix metalloproteinase-14 is a predictor for antifibrotic effect of IC-2-engineered mesenchymal stem cell sheets on liver fibrosis in mice. Regen. Ther. 2021, 26, 292–301. [Google Scholar] [CrossRef]
  35. Duman, D.G.; Zibandeh, N.; Ugurlu, M.U.; Celikel, C.; Akkoc, T.; Banzragch, M.; Genc, D.; Ozdogan, O.; Akkoc, T. Mesenchymal stem cells suppress hepatic fibrosis accompanied by expanded intrahepatic natural killer cells in rat fibrosis model. Mol. Biol. Rep. 2019, 46, 2997–3008. [Google Scholar] [CrossRef]
  36. Bolinas, D.K.M.; Barcena, A.J.R.; Mishra, A.; Bernardino, M.R.; Lin, V.; Heralde, F.M., 3rd; Chintalapani, G.; Fowlkes, N.W.; Huang, S.Y.; Melancon, M.P. Mesenchymal stem cells loaded in injectable alginate hydrogels promote liver growth and attenuate liver fibrosis in cirrhotic rats. Gels 2025, 11, 250. [Google Scholar] [CrossRef]
  37. Shagidulin, M.; Onishchenko, N.; Grechina, A.; Nikolskaya, A.; Krasheninnikov, M.; Lyundup, A.; Volkova, E.; Mogeiko, N.; Venediktov, A.; Piavchenko, G.; et al. Recombinant spidroin microgel as the base of cell-engineered constructs mediates liver regeneration in rats. Polymers 2022, 14, 3179. [Google Scholar] [CrossRef]
  38. Shagidulin, M.; Onishchenko, N.; Sevastianov, V.; Krasheninnikov, M.; Lyundup, A.; Nikolskaya, A.; Kryzhanovskaya, A.; Voznesenskaia, S.; Gorelova, M.; Perova, N.; et al. Experimental correction and treatment of chronic liver failure using implantable cell-engineering constructs of the auxiliary liver based on a bioactive heterogeneous biopolymer hydrogel. Gels 2023, 9, 456. [Google Scholar] [CrossRef]
  39. Onishchenko, N.; Shagidulin, M.; Gonikova, Z.; Nikolskaya, A.; Kirsanova, L.; Venediktov, A.; Pokidova, K.; Kuzmin, E.; Kuznetsova, N.; Kozlov, I.; et al. Hematopoietic stem cells of bone marrow and their total RNA in tat liver regeneration. Appl. Sci. 2025, 15, 3782. [Google Scholar] [CrossRef]
  40. Fotouh, A.; Elbarbary, N.K.; Momenah, M.A.; Khormi, M.A.; Mohamed, W.H.; Sherkawy, H.S.; Ahmed, A.E.; Diab, M.; Elshafae, S. Hepatoprotective effects of mesenchymal stem cells in carbon tetrachloride-induced liver toxicity in rats: Restoration of liver parameters and histopathological evaluation. Am. J. Vet. Res. 2025, 86, 1–10. [Google Scholar] [CrossRef] [PubMed]
  41. Dremin, V.; Potapova, E.; Zherebtsov, E.; Kozlov, I.; Seryogina, E.; Kandurova, K.; Alekseyev, A.; Piavchenko, G.; Kuznetsov, S.; Mamoshin, A.; et al. Optical fine-needle aspiration biopsy in a rat model. In Proceedings of the Dynamics and Fluctuations in Biomedical Photonics XVI, San Francisco, CA, USA, 2–3 February 2019; Volume 10877. [Google Scholar] [CrossRef]
  42. Kuo, W.T.; Hsiao, C.Y.; Chiu, S.H.; Chen, T.H.; Shyu, J.F.; Lin, C.H.; Tsai, P.J. Hypoxia-enhanced Wharton’s jelly mesenchymal stem cell therapy for liver fibrosis: A comparative study in a rat model. Kaohsiung J. Med. Sci. 2025, 41, e70053. [Google Scholar] [CrossRef] [PubMed]
  43. Mo, M.; Wang, S.; Zhou, Y.; Li, H.; Wu, Y. Mesenchymal stem cell subpopulations: Phenotype, property and therapeutic potential. Cell Mol. Life Sci. 2016, 73, 3311–3321. [Google Scholar] [CrossRef]
  44. Nishina, T.; Haga, H.; Wakao, S.; Maki, K.; Mizuno, K.; Katsumi, T.; Hoshikawa, K.T.; Saito, T.; Iseki, M.; Unno, M.; et al. Safety and effectiveness of muse cell transplantation in a large-animal model of hepatic fibrosis. Stem Cells Int. 2025, 2025, 6699571. [Google Scholar] [CrossRef] [PubMed]
  45. Bao, Y.L.; Wang, L.; Pan, H.T.; Zhang, T.R.; Chen, Y.H.; Xu, S.J.; Mao, X.L.; Li, S.W. Animal and organoid models of liver fibrosis. Front. Physiol. 2021, 12, 666138. [Google Scholar] [CrossRef]
  46. Zhang, L.; Liu, C.; Yin, L.; Huang, C.; Fan, S. Mangiferin relieves CCl4-induced liver fibrosis in mice. Sci. Rep. 2023, 13, 4172. [Google Scholar] [CrossRef]
  47. Ding, K.; Liu, M.R.; Li, J.; Huang, K.; Liang, Y.; Shang, X.; Chen, J.; Mu, J.; Liu, H. Establishment of a liver fibrosis model in cynomolgus monkeys. Exp. Toxicol. Pathol. 2014, 66, 257–261. [Google Scholar] [CrossRef]
  48. Enciso, N.; Amiel, J.; Fabián-Domínguez, F.; Pando, J.; Rojas, N.; Cisneros-Huamaní, C.; Nava, E.; Enciso, J. Model of liver fibrosis induction by thioacetamide in rats for regenerative therapy studies. Anal. Cell Pathol. 2022, 2022, 2841894. [Google Scholar] [CrossRef] [PubMed]
  49. Li, W.; Zhu, C.; Li, Y.; Wu, Q.; Gao, R. Mest attenuates CCl4-induced liver fibrosis in rats by inhibiting the Wnt/β-catenin signaling pathway. Gut Liver 2014, 8, 282–291. [Google Scholar] [CrossRef] [PubMed]
  50. Wang, R.; Song, F.; Li, S.; Wu, B.; Gu, Y.; Yuan, Y. Salvianolic acid A attenuates CCl4-induced liver fibrosis by regulating the PI3K/AKT/mTOR, Bcl-2/Bax and caspase-3/cleaved caspase-3 signaling pathways. Drug Des. Devel Ther. 2019, 13, 1889–1900. [Google Scholar] [CrossRef]
  51. Alejolowo, O.O.; Oluba, O.M.; Adeyemi, O.S. Anogeissus leiocarpus (DC.) Guill. & Perr. extract-protected rats against CCl4-induced liver fibrosis. J. Taibah Univ. Med. Sci. 2025, 20, 474–486. [Google Scholar] [CrossRef]
  52. Weber, S.N.; Wasmuth, H.E. Liver fibrosis: From animal models to mapping of human risk variants. Best. Pract. Res. Clin. Gastroenterol. 2010, 24, 635–646. [Google Scholar] [CrossRef]
  53. Lai, C.; Feng, T.; Wei, L.; Zhang, T.; Zhou, G.; Yang, Q.; Lan, T.; Xiang, G.; Yao, Y.; Zhou, L.; et al. Development and validation of a primate model for liver fibrosis. J. Pharmacol. Toxicol. Methods 2019, 100, 106600. [Google Scholar] [CrossRef]
  54. Eom, Y.W.; Shim, K.Y.; Baik, S.K. Mesenchymal stem cell therapy for liver fibrosis. Korean J. Intern. Med. 2015, 30, 580–589. [Google Scholar] [CrossRef] [PubMed]
  55. Li, T.T.; Wang, Z.R.; Yao, W.Q.; Linghu, E.Q.; Wang, F.S.; Shi, L. Stem cell therapies for chronic liver diseases: Progress and challenges. Stem Cells Transl. Med. 2022, 11, 900–911. [Google Scholar] [CrossRef] [PubMed]
  56. Huang, W.C.; Li, Y.C.; Chen, P.X.; Ma, K.S.; Wang, L.T. Mesenchymal stem cell therapy as a game-changer in liver diseases: Review of current clinical trials. Stem Cell Res. Ther. 2025, 16, 3. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Design of the study. (1) Donor animals for obtaining MSCs and experimental animals randomized into study groups; (2) MSC cultivation and further validation of immunological phenotype via flow cytometry; (3) MSC grafting to rats with a CCl4-provoked model of liver fibrosis; (4) blood sampling for assessment of biochemical markers of liver damage and inflammation and histological study of 5 μm liver Mallory-stained sections.
Figure 1. Design of the study. (1) Donor animals for obtaining MSCs and experimental animals randomized into study groups; (2) MSC cultivation and further validation of immunological phenotype via flow cytometry; (3) MSC grafting to rats with a CCl4-provoked model of liver fibrosis; (4) blood sampling for assessment of biochemical markers of liver damage and inflammation and histological study of 5 μm liver Mallory-stained sections.
Applsci 15 10471 g001
Figure 2. Flow cytometry in MSC-treated liver fibrosis in rats. (A). On the left: side-to-forward scattering of signal height (SSC-H/FSC-H) showing a zone that is free of debris. On the right: forward scattering of signal height to area (FSC-H/FSC-A) shows a zone free of doublets; (B) peaks of cell fluorescence with the number of events for the Y-axis and wave intensity for the X-axis.
Figure 2. Flow cytometry in MSC-treated liver fibrosis in rats. (A). On the left: side-to-forward scattering of signal height (SSC-H/FSC-H) showing a zone that is free of debris. On the right: forward scattering of signal height to area (FSC-H/FSC-A) shows a zone free of doublets; (B) peaks of cell fluorescence with the number of events for the Y-axis and wave intensity for the X-axis.
Applsci 15 10471 g002
Figure 3. MSC cultivation for grafting to rats with liver fibrosis. Phase contrast imaging, ×20, scale bar 200 μm: (A) primary culture of MSCs from intact donor rats; (B) first passage of MSCs, phase contrast imaging; (C) second passage of MSCs. Stained slides, ×100, scale bar 100 μm: (D) adipogenic lineage, oil red O staining; (E) osteogenic lineage, alizarin red S staining.
Figure 3. MSC cultivation for grafting to rats with liver fibrosis. Phase contrast imaging, ×20, scale bar 200 μm: (A) primary culture of MSCs from intact donor rats; (B) first passage of MSCs, phase contrast imaging; (C) second passage of MSCs. Stained slides, ×100, scale bar 100 μm: (D) adipogenic lineage, oil red O staining; (E) osteogenic lineage, alizarin red S staining.
Applsci 15 10471 g003
Figure 4. Blood markers in MSC-treated liver fibrosis in rats (n = 24 per group). (A) Timelines for blood levels of liver damage markers, alkaline phosphatase (ALP), alanine aminotransferase (ALT), and aspartate aminotransferase (AST); (B) timelines for blood levels of pro-inflammatory cytokines: interleukins 4 and 10, gamma interferon (IFN-γ), and tumor necrosis factor alpha (TNF-α). Group 1: single event of MSC grafting; Group 2: two events of MSC grafting; Group 3: sham treatment after CCl4. * p < 0.05; ** p < 0.01; *** p < 0.001. Purple asterisks mark differences between Groups 1 and 3; yellow asterisks—between Groups 2 and 3.
Figure 4. Blood markers in MSC-treated liver fibrosis in rats (n = 24 per group). (A) Timelines for blood levels of liver damage markers, alkaline phosphatase (ALP), alanine aminotransferase (ALT), and aspartate aminotransferase (AST); (B) timelines for blood levels of pro-inflammatory cytokines: interleukins 4 and 10, gamma interferon (IFN-γ), and tumor necrosis factor alpha (TNF-α). Group 1: single event of MSC grafting; Group 2: two events of MSC grafting; Group 3: sham treatment after CCl4. * p < 0.05; ** p < 0.01; *** p < 0.001. Purple asterisks mark differences between Groups 1 and 3; yellow asterisks—between Groups 2 and 3.
Applsci 15 10471 g004
Figure 5. Histological study of liver sections for MSC-treated liver fibrosis in rats (n = 24 per group). (A) 5 μm sections, Mallory’s staining, ×40, scale bar 100 μm; (B) timelines for the number of false lobules per field of view; (C) timelines for the area of connective tissue proper per field of view; (D) timelines for the number of hepatocytes with two nuclei per field of view. Group 1: single event of MSC grafting; Group 2: double event of MSC grafting; Group 3: no treatment. Red arrows mark the zones of perilobular degeneration, appearance of adipocytes, degraded structure of hepatic plates, and areas of blood congestion in widened sinusoidal capillaries between the plates. Black arrows mark binucleated hepatocytes. Blue arrows mark the stromal component consisting of loose connective tissue.
Figure 5. Histological study of liver sections for MSC-treated liver fibrosis in rats (n = 24 per group). (A) 5 μm sections, Mallory’s staining, ×40, scale bar 100 μm; (B) timelines for the number of false lobules per field of view; (C) timelines for the area of connective tissue proper per field of view; (D) timelines for the number of hepatocytes with two nuclei per field of view. Group 1: single event of MSC grafting; Group 2: double event of MSC grafting; Group 3: no treatment. Red arrows mark the zones of perilobular degeneration, appearance of adipocytes, degraded structure of hepatic plates, and areas of blood congestion in widened sinusoidal capillaries between the plates. Black arrows mark binucleated hepatocytes. Blue arrows mark the stromal component consisting of loose connective tissue.
Applsci 15 10471 g005
Figure 6. Immunofluorescent study of liver sections for MSC-treated liver fibrosis in rats (n = 24 per group). (A) 5 μm sections, anti-Ki67 staining and DAPI, ×40, scale bar 100 μm. Red arrows mark the zones of dystrophic changes. White arrows mark Ki67-positive cells. (B) plot showing the number of Ki67 cells per field of view. Group 1: single event of MSC grafting; Group 2: double event of MSC grafting; Group 3: no treatment of liver fibrosis.
Figure 6. Immunofluorescent study of liver sections for MSC-treated liver fibrosis in rats (n = 24 per group). (A) 5 μm sections, anti-Ki67 staining and DAPI, ×40, scale bar 100 μm. Red arrows mark the zones of dystrophic changes. White arrows mark Ki67-positive cells. (B) plot showing the number of Ki67 cells per field of view. Group 1: single event of MSC grafting; Group 2: double event of MSC grafting; Group 3: no treatment of liver fibrosis.
Applsci 15 10471 g006
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

Lyundup, A.; Shagidulin, M.; Onishchenko, N.; Beregovykh, V.; Krasheninnikov, M.; Venediktov, A.; Pokidova, K.; Nikolskaya, A.; Kuzmin, E.; Kostin, A.; et al. Mesenchymal Stem Cells in Liver Fibrosis: A Dose-Dependent Recovery. Appl. Sci. 2025, 15, 10471. https://doi.org/10.3390/app151910471

AMA Style

Lyundup A, Shagidulin M, Onishchenko N, Beregovykh V, Krasheninnikov M, Venediktov A, Pokidova K, Nikolskaya A, Kuzmin E, Kostin A, et al. Mesenchymal Stem Cells in Liver Fibrosis: A Dose-Dependent Recovery. Applied Sciences. 2025; 15(19):10471. https://doi.org/10.3390/app151910471

Chicago/Turabian Style

Lyundup, Aleksey, Murat Shagidulin, Nina Onishchenko, Valery Beregovykh, Mikhail Krasheninnikov, Artem Venediktov, Ksenia Pokidova, Alla Nikolskaya, Egor Kuzmin, Andrey Kostin, and et al. 2025. "Mesenchymal Stem Cells in Liver Fibrosis: A Dose-Dependent Recovery" Applied Sciences 15, no. 19: 10471. https://doi.org/10.3390/app151910471

APA Style

Lyundup, A., Shagidulin, M., Onishchenko, N., Beregovykh, V., Krasheninnikov, M., Venediktov, A., Pokidova, K., Nikolskaya, A., Kuzmin, E., Kostin, A., Arzhanova, A., Fadeev, P., Kuznetsova, N., Piavchenko, G., & Gautier, S. (2025). Mesenchymal Stem Cells in Liver Fibrosis: A Dose-Dependent Recovery. Applied Sciences, 15(19), 10471. https://doi.org/10.3390/app151910471

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