Loading

Archives of Stem Cell and Research

Cyclosporin a Inhibits Adipogenic Differentiation through the TGF- ?1 Signaling Pathway and Regulates Immunomodulatory Functions of Mesenchymal Stem Cells in Aplastic Anemia Patients

Research Article | Open Access | Volume 6 | Issue 1

  • 1. Department of Hematology, Zhengzhou central hospital affiliated Zhengzhou University, Zhengzhou, China
  • 2. Center of stem cell and regenerative medicine, Zhengzhou central hospital affiliated Zhengzhou University, Zhengzhou, China
  • 3. Stem Cell Research Center, Henan key Laboratory of stem cell Differentiation and Modification Henan Provincial Peoples Hospital, Peoples Hospital of Zhengzhou University, Zhengzhou, China
+ Show More - Show Less
Corresponding Authors
Han Yue, Stem Cell Research Center, Henan key Laboratory of stem cell Differentiation and Modification Henan Provincial Peoples Hospital, Peoples Hospital of Zhengzhou University, Zhengzhou, China
Abstract

Aplastic Anemia (AA) is a syndrome of bone marrow hematopoietic failure manifested also by pancytopenia, hemorrhage, and infection. The pathogenesis of AA involves Bone Marrow-Derived Mesenchymal Stem Cells (BM-MSCs), which are key components of the bone marrow hematopoietic microenvironment and hematopoietic regulation. Abnormal activation of the immune system plays an important role in the pathogenesis of aplastic anemia. The immunosuppressant Cyclosporine (CsA) is widely used to treat AA, but its effect on BM-MSCs remains unclear. In this study, the effects of CsA on proliferation, apoptosis, morphology, matrilineage differentiation and immune regulation of BM-MSCs in AA patients were verified. Our results show that CsA regulates the bone marrow microenvironment in AA by reducing BM-MSCs adipogenic differentiation through the transforming growth factor beta 1 signaling pathway. Meanwhile, CsA can upregulate the expression levels of PD-L1 and PD-L2, especially PD-L1 positive cells, by transforming growth factor beta secretion and/or reducing the levels of IL6, further enhance the immunosuppressive ability of BM-MSCs which is beneficial for treating AA.

Keywords

• Cyclosporin A

• Aplastic Anemia

• Mesenchymal Stem Cell

• Immunological Regulation

• Signaling Pathway

• Programmed Cell Death Protein 1

CITATION

Shi LF, Sun L, Ma BD, Guo ZQ, Jin RR, et al. (2024) Cyclosporin a Inhibits Adipogenic Differentiation through the TGF- β1 Signaling Pathway and Regulates Immunomodulatory Functions of Mesenchymal Stem Cells in Aplastic Anemia Patients. Arch Stem Cell Res 6(1): 1020.

INTRODUCTION

Aplastic Anemia (AA) is a syndrome affecting hematopoietic Stem Cells and their microenvironment and is associated with bone marrow tissue lipogenesis, abnormal function, and pancytopenia [1] as well as infection and hemorrhage. The latter clinical manifestations are mediated by aberrant immune cells [2]. Which directly and indirectly destroy stem/progenitor cells in the bone marrow by secreting gamma interferon, tumor necrosis factor alpha, and interleukins. Meanwhile, hematopoiesis is suppressed as a result of decreased production of early hematopoietic growth factors by stromal cells [3]. In addition to the sharp decline in the number of hematopoietic cells in the bone marrow of patients with AA, there is a dramatic increase in the number fat cells [4]. As lipogenesis is a notable pathological feature of AA, it is possible that this is related to abnormal differentiation of stem cells in bone marrow.

Bone Marrow-Derived Mesenchymal Stem Cells (BM-MSCs) self-renew, proliferate, and differentiate into a variety of cell types, including neurons, glia, and endothelial cells, as well as adipose and cartilage [5,6] and have therefore attracted considerable attention in the field of tissue regeneration. MSCs help maintain hemopoiesis and exert immune regulation, but are altered in patients with a variety of hematological diseases. BM-MSCs from patients with AA have enhanced angiogenesis as well as intrinsic deficits that contribute to their vulnerability to bone marrow failure. In addition, BM-MSCs have negative immunomodulatory properties, secreting cytokines to regulate T and B-lymphocytes, NK cells, and dendritic cells. MSCs express cell surface molecules with an immunosuppressive capacity, such as Programmed Death Ligand 1 (PD-L1) [7].

Lipogenesis is one of the most significant pathological features of bone marrow in aplastic anemia patients. The number of hematopoietic cells in the bone marrow of aplastic anemia patients decreases sharply while the number of fat cells increases significantly [8]. Obviously, these fat cells originate from the lipogenesis of bone marrow cells. Therefore, we speculated that the advocates in the bone marrow cavity of aplastic anemia patients might be related to the abnormal differentiation potential of MSC in aplastic anemia patients.

Current standard regimens for acquired AA include antilymphocyte/antithymocyte globulin and cyclosporine (or Cyclosporin A [CsA]), which target overactivated T cells [9,10], or marrow transplantation. Antilymphocyte/antithymocyte globulin selectively binds T lymphocytes and, with serum complement, lyses peripheral blood lymphocytes and reduces the expression of Fas antigen in CD34 + cells in bone marrow. At the same time, antithymocyte globulin also promotes the secretion of hematopoietic cell growth factor from lymphocytes. CsA, an immunosuppressant that is commonly used to treat organ transplant rejection and autoimmune disease as well as AA [11,12], inhibits the production of IL-1 by macrophages and the expression of the IL-1 receptor on Th cells and indirectly enhances the growth of granular monocytes. However, the effect of CsA on human BM-MSCs is not known. The objective of this study was to assess whether CsA affects proliferation, apoptosis, and immunomodulatory functions of human BM-MSCs. Furthermore, this study explored the role of CsA in lipogenesis as a novel mechanism for the therapeutic effects of CsA on BM-MSCs in patients with AA.

MATERIALS AND METHOD

Culture of Human BM-Mscs

Three patients diagnosed with AA according to the criteria of The Fourth National Aplastic Anemia Conference (1987) at the blood department of Zhengzhou Central Hospital affiliated to Zhengzhou University were included in the study. Three marrow samples were obtained from the ribs that were removed from bone marrow puncture. Patients were informed of the content of the study and voluntarily acceded to participate. All donors provided written informed consent, and the Ethics Committee of Zhengzhou Central Hospital Affiliated to Zhengzhou University approved the present study. Those who had recently received related drug treatment, had contraindications to CsA, autoimmune diseases, or malignant tumors were excluded from the study.

BM-MSCs were extracted from patient samples named AA- BM-MSCs and cultured in Dulbecco’s modified Eagle medium/ F12 supplemented with 10% fetal bovine serum, 100 IU/ml penicillin, and 100 μg/ml streptomycin. Only BM-MSCs passaged 1 to 3 times were used in the study. When cells reached semi confluence, culture plates were washed and the MSCs phenotype confirmed by flow cytometry (Cytoflex, Beckman Coulter) using the specific antibodies anti-CD73-PE, anti-CD90-APC, anti- CD105-APC-A750, anti-CD34, anti-CD45, anti-CD11b, anti-CD19, and anti-HLA-DR-FITC (Biolegend, San Diego, CA USA).

Cell Proliferation

Here AA-BM-MSCs were treated with the drug on two groups, which respectively named CsA and NC (DMSO negative control). AA-BM-MSCs were seeded at a density of 7×103 cells per well in 96-well culture plates and treated with CsA (Novartis Pharma AG, Basel, Switzerland) at concentrations of 0, 0.5, 1, 2, 5, and 10 μg/ ml for 24 h. Cell proliferation was measured by the CCK8 assay (Beyotime, Shanghai, China), and the absorbance was measured at 450 nm using a microplate reader (BioTek Instruments, Inc. Winooski, VT, USA).

Cell Apoptosis

AA-BM-MSCs were plated at a density of 7×104 cells per well in 6-well culture plates and treated with CsA (0-10 μg/ml) for 24 h. Apoptosis was assessed by staining with annexin V/fluorescein isothiocyanate and propidium iodide and analyzed using a Beckman flow cytometer (Beckman Coulter, Brea, CA, USA).

Flow Cytometry Analysis

Cells were resuspended in 500 μl PBS with antibodies against anti-CD73-PE, anti-CD90-APC, anti-CD105-APC-A750, anti-CD34, anti-CD45, anti-CD11b, anti-CD19, anti-HLA-DR-FITC, anti-PDL1- FITC, and anti-PDL2-PE (all from BioLegend, USA) at 4°C for 30 min. The samples were washed twice, and analyzed using a Beckman flow cytometer (Beckman Coulter, Pasadena, CA).

AA-BM-MSCs Adipocyte Differentiation and Osteogenic Differentiation

To induce adipocyte or osteogenic differentiation, AA-BM- MSCs were plated at a density of 3×104 cells per well in 6-well plates and treated with CsA at 2 μg/mL for 72 h (we found CsA almost had no effect on cell proliferation and apoptosis at this concentration). For some experiments, cells were pretreated with 5 nM A8301 (an inhibitor of transforming growth factor beta 1 receptor kinases ALK4, -5, and -7) for 1 h before CsA was applied. Osteogenic differentiation medium (Lonza) containing L-glutamine, mesenchymal cell growth serum MCGS, dexamethasone, ascorbate, β-glycerophosphate, and pen/strep. Adipogenic induction medium (Lonza) containing additional h-insulin, L-glutamine, MCGS, dexamethasone, indomethacin, 3-isobuty-1-methyl-xanthine, and pen/strep. Cell culture medium was changed every 2-3 days for a total of 21 days. On day 21, Oil Red O solution was used to stain the accumulated lipid droplets in differentiated adipocytes observed by microscopy. Alizarin red staining (Solarbio, Beijing, China) was performed to assess osteogenic differentiation.

RNA Isolation and qPCR

Total RNA was isolated from AA-BM-MSCs for real-time PCR (qPCR) using the primers listed in (Table 1). Gene expression was normalized to β-actin levels.

Table 1: Primer sequences used in this study.

Gene

Primer

 

 

 

Direction Sequence (5’- 3’)

 

Length (bp)

 

β-Actin

Forward

CATCCGTAAAGACCTCTATGCCAAC

 

171

Reverse

ATGGAGCCACCGATCCACA

 

Ocn

Forward

AGCAGCTTGGCCCAGACCTA

 

178

Reverse

TAGCGCCGGAGTCTGTTCACTAC

 

Co11a1

Forward

GAAGTCAGCTGCATACAC

 

312

Reverse

AGGAAGTCCAGGCTGTCC

 

PPAR-γ

Forward

TGTCGGTTTCAGAAGTGCCTTG

 

122

Reverse

TTCAGCTGGTCGATATCACTGGAG

 

Runx2

Forward

TGGCTCAGATAAGAGGGGTAAGA

 

257

Reverse

GAACCAAGAAGGCACAGACAGAA

 

Bcl-2

Forward

TACCGTCGTGACTTCGCAGAG

 

350

Reverse

GGCAGGCTGAGCAGGGTCTT

 

Bax

Forward

CGGCGAATTGGAGATGAACTG

 

160

Reverse

GCAAAGTAGAAGAGGGCAACC

 

PD-L1

Forward

AAAGTCAATGCCCCATACCG

 

151

Reverse

TTCTCTTCCCACTCACGGGT

 

PD-L2

Forward

CGTGACAGCCCCTAAAGAAG

 

250

Reverse

GATGACCAGGCAACGGTACT

Enzyme-Linked Immunosorbent Assay (ELISA)

AA-BM-MSCs were stimulated with CsA at 2 μg/ml for 72 h. Secretion of transforming growth factor beta (TGF-β) was measured using ELISA as per the manufacturer’s instructions (R&D Systems, Minneapolis, MN, USA).

Flow High Flux Multifactor Detection

The supernatants of AA-BM-MSCs+CsA co-cultured for 24 h were collected and Legendplex TM human Th cytokine assay (Biolegend) was used to analyze the following 13 factors: IL-2, 4, 5, 6, 9, 10, 13, 17A17F, 21, 22, IFN-γ and TNF-α. Experiments were repeated to ensure the accuracy of the data. Finally, Cytoflex flow cytometer (Beckman Coulter, Krefeld, USA) was used for analysis and Legendplex V8.0 and Prism7.0 software (Biolegend) analyzed the data, the concentration was ng/ml.

Statistical Analysis

Statistical analyses were performed using Prism 7.0 (GraphPad Software, San Diego, CA) SPSS professional statistical software. The experimental results are reported as means ± standard deviations. The Student t test was used to compare two groups, and one-way analysis of variance was used for comparisons among groups. A P value of < 0.05 was considered statistically significant.

RESULTS

CsA Decreased AA-BM-Mscs Proliferation and Increased Cell Apoptosis

To investigate whether CsA affects AA-BM-MSC proliferation and apoptosis, cells were treated with CsA at concentrations of 0, 0.5, 1, 2, 5, and 10 μg/ml for 24 h. CCK-8 assays showed that CsA significantly inhibited AA-BM-MSC proliferation at 5 μg/ml and 10 μg/ml (Figure 1A).

Effects of CsA on the proliferation and apoptosis of AA-BM-MSCs. A. Cell proliferation of BM-MSCs treated with CsA (0-10 ?g/ml) was assayed by CCK8.  B. The mRNA expression of BCL2 and BAX in BM-MSCs was determined by real-time PCR.  C,D. CsA promoted cell apoptosis in vitro according to annexin V-FITC/propidium iodide double staining and flow cytometry. ***P < 0.001.

Figure 1: Effects of CsA on the proliferation and apoptosis of AA-BM-MSCs.

A. Cell proliferation of BM-MSCs treated with CsA (0-10 μg/ml) was assayed by CCK8.

B. The mRNA expression of BCL2 and BAX in BM-MSCs was determined by real-time PCR.

C,D. CsA promoted cell apoptosis in vitro according to annexin V-FITC/propidium iodide double staining and flow cytometry. ***P < 0.001.

This concentration also reduced the ratio of antiapoptotic Bcl-2 gene expression to that of proapoptotic Bax according to qPCR (Figure 1B). Accordingly, annexin V/ fluorescein isothiocyanate and propidium iodide staining revealed that 5 μg/ml and 10 μg/ml CsA increased the percentage of cells undergoing apoptosis (Figure 1C,D). Thus, CsA promotes both early and late phases of apoptosis.

CsA Had No Effect on Immunophenotype and Osteogenic Differentiation of AA-BM-Mscs

To determine whether CsA alters the immunophenotype of AA-BM-MSCs, the expression of key surface markers was tested by flow cytometry, such as (Figure 2A).

Effects of CsA on surface markers and osteogenic differentiation of AA-BM-MSCs. A. Flow cytometry for surface markers of human BM-MSCs treated or not with 2 ?g/ml CsA. The histogram shows the relative expression of each factor. B. The osteogenic mRNA expression of Ocn, Col1a1 and Runx2 was determined in 3d, 13d and 21d. C. Osteogenic differentiation of BM-MSCs was illustrated by Alizarin red staining

Figure 2: Effects of CsA on surface markers and osteogenic differentiation of AA-BM-MSCs.

A. Flow cytometry for surface markers of human BM-MSCs treated or not with 2 μg/ml CsA. The histogram shows the relative expression of each factor.

B. The osteogenic mRNA expression of Ocn, Col1a1 and Runx2 was determined in 3d, 13d and 21d.

C. Osteogenic differentiation of BM-MSCs was illustrated by Alizarin red staining

However, the expression of these cell-surface markers was unaffected by treatment for 72 h with 2 μg/ml CsA. After osteogenic induction for 3, 13 and 21 days, the osteogenic gene expression and Alizarin red staining in BM-MSCs were observed. We found that CsA did not affect the mRNA expression of OCN, COL1a1 and Runx2 at 2 μg/ml (Figure 2B). Three weeks after osteogenic induction, we found no evidence that CsA could promote the osteogenic differentiation of AA-BM-MSCs, as illustrated by Alizarin red staining (Figure 2C).

CsA Inhibited The Lipogenic Differentiation Of AA- BM-Mscs Through The TGF- β1 Signaling Pathway

AA-BM-MSCs were treated with CsA for 3 days at 2 μg/ml and were cultured in appropriate conditions for inducing the adipogenic differentiation. Our results indicated that the mRNA expressions of PPAR-γ was significantly decreased in CsA-treated AA-BM-MSCs (Figure 3A).

 Effect of CsA on lipogenic differentiation and TGF-? concentrations of AA-BM-MSCs.  A,D. Oil-Red O stainings of CsA-treated, CsA+A8301-treated AA-BM-MSCs and controls after cultured in adipogenic medium for 21 days. B,E. The mRNA expression of adipogenic PPAR-? was determined by real time PCR.  C. TGF-? concentrations were measured by enzyme-linked immunosorbent assays.***P < 0.001.

Figure 3: Effect of CsA on lipogenic differentiation and TGF-β concentrations of AA-BM-MSCs.

A,D. Oil-Red O stainings of CsA-treated, CsA+A8301-treated AA-BM-MSCs and controls after cultured in adipogenic medium for 21 days.

B,E. The mRNA expression of adipogenic PPAR-γ was determined by real time PCR.

C. TGF-β concentrations were measured by enzyme-linked immunosorbent assays.***P < 0.001.

Oil Red-O staining results showed that CsA inhibited the lipogenic differentiation of AA-BM-MSCs (Figure 3B). It is widely assumed that TGF-β exerts an inhibitory action on human adipose tissue. Then we analyzed TGF-β secretion by ELISA. Our results showed that the secretion level of TGF-β in the supernatant was considerably increased with CsA treatment (Figure 3C).

To further confirm the inhibitory effect of CsA on the adipogenic differentiation of AA-BM-MSCs, we pretreated the cells with TGF-β1 signaling pathway block-blocking agent A8301 (5 nM), and detected the lipid differentiation ability, expression level of mRNA of PPAR-γ. Our results showed that lipotropic ability was significantly up regulated after induction by the lipotropic medium (Figure 3D). At the same time, the mRNA expression level of PPAR-γ was also significantly up-regulated (Figure 3E). These results suggest that CsA mainly inhibits the lipogenic differentiation of AA-BM-MSCs in vitro through the TGF-β1 signaling pathway.

CsA Treatment Inhibited AA-BM-Mscs Secreting IL-6 And Upregulated The Expression Of PD-L1 And PD-L2

As we know, CsA is an immunosuppressant that selectively acts on the CD4 + subset of T lymphocytes. So we determine the effect of CsA on the immunomodulation function of AA-BM-MSCs, we analyzed cytokine production via flow high-flux multifactor detection. This panel allows simultaneous quantification of 13 human cytokines, including IL-2, 4, 5, 6, 9, 10, 13, 17A, 17F, 21,22, IFN-γ and TNF-α, which are collectively secreted by Th1, Th2, Th9, Th17, and Th22. Medium from the CsA-treated AA-BM-MSCs had lower concentrations of IL-13, and IFN-γ than NC, and the secretion level of IL-6 was significantly reduced (Figure 4A).

 Effect of CsA on the immunomodulation function of AA-BM-MSCs.  A. IL-2, 4, 5, 6, 9, 10, 13, 17A, 17F, 21, 22, IFN-? and TNF-? were detected by Human the Cytokine Panel. B. mRNA expression levels of PD-L1 and PD-L2 in human AA-BM-MSCs treated or not with CsA. C. Surface expression of PD-L1 were assessed by flow cytometry. D. Quantitative analysis of PD-L1 positive cells from three independent experiments. E. Surface expression of PD-L2 were assessed by flow cytometry.  F. Quantitative analysis of PD-L2 positive cells from three independent experiments.*P < 0.05, ***P < 0.001.

Figure 4: Effect of CsA on the immunomodulation function of AA-BM-MSCs.

A. IL-2, 4, 5, 6, 9, 10, 13, 17A, 17F, 21, 22, IFN-γ and TNF-α were detected by Human the Cytokine Panel.

B. mRNA expression levels of PD-L1 and PD-L2 in human AA-BM-MSCs treated or not with CsA.

C. Surface expression of PD-L1 were assessed by flow cytometry.

D. Quantitative analysis of PD-L1 positive cells from three independent experiments.

E. Surface expression of PD-L2 were assessed by flow cytometry.

F. Quantitative analysis of PD-L2 positive cells from three independent experiments.*P < 0.05, ***P < 0.001.

As previously reported, TGF-β1 and IL-6 signaling can regulate the expression of PD-L1 and PD-L2 [13,14]. Therefore, we verified the expression of PD-L1 and PD-L2. CsA treatment upregulated the expression of PD-L1 and PD-L2 mRNA in AA- BM-MSCs (Figure 4B). Flow cytometry analyses revealed that protein expression of PD-L1 and PD-L2 on CsA-treated AA-BM- MSCs was all higher than controls (Figure 4C,E). Interestingly, almost all (95.74% ± 3.25%) AA-BM-MSCs expressed PD-L2 (Figure 4F). However, CsA treatment increased the percentage of PD-L1 positive cells from 9.98% ± 2.47% to 58.32% ± 2.85% (Figure 4D).

DISCUSSION

Cyclosporine (CsA) is a traditional immunosuppressant that is commonly used to treat organ transplant rejection, Aplastic Anemia (AA) and autoimmune diseases [15,16]. BM-MSCs are the main component of the bone marrow microenvironment.

The main function of MSCs is to maintain hemopoiesis and exert immune regulation [17,18]. But they can also differentiate into adipose, osteogenic, and cartilage tissues and secrete a variety of factors. MSCs are critically altered in patients with different hematological diseases [19,20]. For example, BM-MSCs from patients with AA enhance angiogenesis, which may play a role in the pathogenesis of the disease [21]. Qu, et al [22] showed that the proliferation and adipogenic differentiation of murine BM-MSCs are inhibited by CsA, which also promotes their apoptosis. Here, we show that CsA affects the proliferation, apoptosis, phenotype, multidirectional differentiation ability, and cytokine secretion of human BM-MSCs from AA patients.

In our study, we confirmed that CSA inhibits BM-MSC proliferation and promotes apoptosis in a dose-dependent manner. Although the mechanism behind the ant proliferative effect remains unclear, CsA has been shown to impair proliferation and osteoclast differentiation via the nitric oxide synthase pathway [23]. CsA-mediated apoptosis, however, likely involved the decrease in the ratio of Bcl-2, one of the most important regulators of apoptosis in mammalian cells, to Bax, a molecule that initiates mitochondrial apoptosis pathways [24,25]. This suggests that Bax and Bcl-2 are involved in CsA inducing apoptosis of BM-MSCs.

Surface antigen expression of MSCs includes (CD73, CD90 and CD105). BM-MSCs have no or low expression of co- stimulatory molecules CD19, CD34, CD45, CD11b and HLA-DR, which contribute to low immunogenicity of BM-MSCs. In our study, we found that CsA did not affect the immunophenotype of BM-MSCs. Fatty Bone Marrow (BM) and defective hematopoiesis are a pathologic hallmark of Aplastic Anemia (AA). Existing literature reports that BM-MSC of AA patients had a morphology, phenotype, and osteogenic differentiation potential similar to normal people but adiposity differentiated from AA BM-MSC had a higher density and larger size of lipid droplets and they expressed significantly higher levels of adiponectin and FABP4 genes and proteins as compared to control BM-MSC [26,27]. AA-BM-MSC have enhanced angiogenesis, which may have an important implication in the pathogenesis of the disease. These studies indicate that BM-MSCs may be associated with the pathogenesis of AA through regulating the microenvironment and inhibiting adipogenesis may be a new therapeutic method for AA. In this study, we investigated the effect of CsA on adipogenic and osteogenic differentiation in BM-MSCs.

In addition, we also examined the lipogenic and osteogenic genes of bone marrow mesenchyme stem cells in aplastic anemia patients with or without CsA treatment CsA inhibited BM-MSCs adipogenic differentiation obviously, while no significant effect on osteogenic differentiation. At the same time, we investigated the changes of cytokines and found that the secretion level of TGF-β in BM-MSCs significantly increased after CsA treatment. We speculated whether CsA was involved in the lipid differentiation of BM-MSCs through the TGF-β pathway. In addition, Gilson [28,29] pointed out that the decreased concentration of TGF-β1 in the blood circulation of aplastic anemia patients was significantly correlated with the occurrence of aplastic anemia. TGF-β1 is an important regulator involved in the adipogenic differentiation of BM-MSCs, and its specific regulatory mechanism remains unclear. In recent years, some progress has been made in its research. Tsurutani, et al [30] reported that TGF-β plays an important role in regulating adipose differentiation of mouse embryonic fibroblasts, but the mechanism is not clear. Mu, et al [31] found that TGF-β can exert multiple biological effects in stem cells through Smad signaling pathways and non-smad signaling pathways. Then we did the TGF-β1 pathway inhibition test. Our results showed that lipotropic ability was significantly up- regulated after induction by the lipotropic medium. At the same time, the mRNA expression level of PPAR-γ was also significantly up-regulated. These results showed that CsA could regulate the bone marrow microenvironment by affecting BM-MSCs through the TGF-β1 signaling pathway, thus promoting the treatment of AA.

Several soluble factors produced by MSCs have been reported to mediate the suppression of T-cell proliferation [32]. IL-6 is a cytokine synthesized and secreted by mononuclear phagocytes and vascular endothelial cells, which are involved in the regulation of immune system function and hematopoietic function of bone marrow tissue [33,34]. Gupta et al found that IL-6 is involved in the severity of children with aplastic anemia [35]. Earlier studies found that IL-6 aggravates the condition of patients with aplastic anemia [36]. The application of immunosuppressive agents at high levels of IL-6 in children with aplastic anemia is effective [37]. However, whether it has a role in aplastic anemia needs further research. Reported that CsA acts selectively on T lymphocyte subsets. Namely, it inhibits the activation and proliferation of T cells by blocking expression of the IL-2 receptor and suppressing the production of IL-2 and IFN-γ. Our study confirmed that CsA may inhibit the secretion of IL-6, IL-13 and IFN-γ by AA bone marrow mesenchyme stem cells to regulate the immune function of AA patients. And then achieve the effect of treating AA. Our results suggested that CsA can inhibit T cell activation, also can improve the bone marrow microenvironment of AA by acting on BM-MSCs. This is consistent with Qu, et al [22] research in murine BM-MSCs.

Previous study showed that BM-MSCs inhibited lymphocyte proliferation via engagement of the inhibitory molecule programmed death-1 (PD-1) to its ligands PD-L1 and PD-L2 [38- 40]. Fresh reports of aplastic anemia as immune-related adverse events (irAEs) implicate PD-1/PD-L1 as important in preventing immune-mediated destruction of the hematopoietic niche [41]. David JM, et al [42] showed that TGF-β was found to up-regulate PD-L1 gene transcription in a Smad2-dependent manner in non-small cell lung cancer cells. Our study discovered that CsA may significantly promote the secretion of TGF-β1 by AA bone marrow mesenchyme stem cells. These findings demonstrate that up-regulation of BM-MSCs PD-L1 is a novel mechanism of TGF-β-induced immunosuppression in CsA treated AA.

We also found CsA significantly inhibits the secretion of IL-6 by AA bone marrow mesenchyme stem cells. Gupta, et al found that IL-6 is involved in the severity of children with aplastic anemia [42]. Earlier studies found that IL-6 aggravates the condition of patients with aplastic anemia. The application of immunosuppressive agents at high levels of IL-6 in children with aplastic anemia is effective [37]. Mace, Hirotake T, et al [14] showed that targeted inhibition of IL-6 may enhance the efficacy of anti-PD-L1 in pancreatic ductal adenocarcinoma. Fang Xu, et al [14] showed that PD-L1 Mesenchymal Stem Cell (MSC) derived extracellular vesicles (MSCs-EVs-PD-L1) exhibits an impressive ability to regulate various activated immune cells to an immunosuppressed state in vitro. Hence, our results indicate a novel mechanism for CsA influencing BM-MSCs. In the treatment of AA with CsA, blocking the IL-6 pathway can help to up-regulate the expression of PD-L1 and enhance the immunosuppressive effect of BM-MSCs to enhance therapeutic effect.

CONCLUSION

Our date showed that CsA inhibited AA-BM-MSC proliferation, while promoting AA-BM-MSCs apoptosis. In addition, CsA induced adipogenic differentiation, but had no effect on osteogenic differentiation in AA-BM-MSCs. We found that higer concentration of TGF-β was detected in the CsA-treated AA-BM- MSCs medium than NC. In addition, we also found that CsA could regulate the bone marrow microenvironment by affecting AA-BM- MSCs adipogenic differentiation through the TGF-β1 signaling pathway. Thus, in the treatment of AA with CsA, blockade of the IL-6 pathway may facilitate PD-L1 expression in AA-BM-MSCs to enhance the therapeutic immunosuppressive effect, which promote the treatment of AA. According to the above results, we will pay attention to the changes of the immune regulatory function of the AA-BM-MSCs on T cell subsets in patients with highly expressed PD-L1 aplastic anemia in the future work.

AUTHORS’ CONTRIBUTIONS

All listed authors have made substantial contributions to the following aspects of the manuscript:

  1. The conception and design of the study, or acquisition of data, or analysis and interpretation of data.
  2. Drafting the article or revising it critically for important intellectual content.
  3. Final approval of the version to be submitted.

Ethics Approval and Consent to Participate: Not applicable.

Human and Animal Rights: No Animals/Humans were used for studies that are base of this research.

Consent for Publication: Not applicable.

Availability of Data and Materials: The data that support the findings of this study are available on request from the corresponding author.

Funding: This work was supported by Henan Province science and technology research and development projects (grant numbers 222102310032).This work was also supported by the Medical science and Technology project of Henan Province (grant numbers LHGJ20200764).This work was also supported by the Medical science and Technology project of Henan Province (grant numbers LHGJ20210775).

REFERENCES
  1. Katagiri T, Kawamoto H, Nakakuki T, Ishiyama K, Okada-Hatakeyama M, Ohtake S, et al. Individual hematopoietic stem cells in human bone marrow of patients with aplastic anemia or myelodysplastic syndrome stably give rise to limited cell lineages. Stem Cells. 2013; 31: 536-546. doi: 10.1002/stem.1301. PMID: 23316019.
  2. Young NS, Calado RT, Scheinberg P. Current concepts in the pathophysiology and treatment of aplastic anemia. Blood. 2006; 108: 2509-2519. doi: 10.1182/blood-2006-03-010777. Epub 2006 Jun 15. PMID: 16778145; PMCID: PMC1895575.
  3. Kovtonyuk LV, Fritsch K, Feng X, Manz MG, Takizawa H. Inflamm-Aging of Hematopoiesis, Hematopoietic Stem Cells, and the Bone Marrow Microenvironment. Front Immunol. 2016; 7: 502. doi: 10.3389/ fimmu.2016.00502. PMID: 27895645; PMCID: PMC5107568.
  4. Katayama Y. [Bone and Stem Cells. Regulation of hematopoietic microenvironment by bone metabolism]. Clin Calcium. 2014; 24: 547-554. Japanese. PMID: 24681501.
  5. Shu XC, Zhu DH, Liu JJ, Yin DC, Pang TJ, Lu HY, et al. [In vitro studies on the growth and proliferation characteristics of rat bone mesenchymal stem cells]. Zhonghua Yu Fang Yi Xue Za Zhi. 2010; 44: 923-927. Chinese. PMID: 21176525.
  6. Hu JB, Zhou Y, Jiang DD, Tan WS. [Proliferation and differentiation characteristics of human bone marrow mesenchymal stem cells during ex-vivo expansion]. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi. 2006; 22: 7-10. Chinese. PMID: 16388733.
  7. Augello A, Tasso R, Negrini SM, Amateis A, Indiveri F, Cancedda R, et al. Bone marrow mesenchymal progenitor cells inhibit lymphocyte proliferation by activation of the programmed death 1 pathway. Eur J Immunol. 2005; 35: 1482-1490. doi: 10.1002/eji.200425405. PMID:15827960.
  8. Frickhofen N, Kaltwasser JP, Schrezenmeier H, Raghavachar A, Vogt HG, Herrmann F, et al. Treatment of aplastic anemia with antilymphocyte globulin and methylprednisolone with or without cyclosporine. The German Aplastic Anemia Study Group. N Engl J Med. 1991; 324: 1297-1304. doi: 10.1056/NEJM199105093241901. PMID: 2017225.
  9. Bacigalupo A, Broccia G, Corda G, Arcese W, Carotenuto M, Gallamini A, et al. Antilymphocyte globulin, cyclosporin, and granulocyte colony-stimulating factor in patients with acquired severe aplastic anemia (SAA): a pilot study of the EBMT SAA Working Party. Blood. 1995; 85: 1348-1353. PMID: 7532040.
  10. Marsh JC, Zomas A, Hows JM, Chapple M, Gordon-Smith EC. Avascular necrosis after treatment of aplastic anaemia with antilymphocyte globulin and high-dose methylprednisolone. Br J Haematol. 1993; 84: 731-735. doi: 10.1111/j.1365-2141.1993.tb03153.x. PMID: 8217834.
  11. Hirose Y, Masaki Y, Ebata K, Okada J, Kim CG, Ogawa N, et al. T-Cell type acute lymphoblastic leukemia following cyclosporin A therapy for aplastic anemia. Int J Hematol. 2001; 73: 226-229. doi: 10.1007/ BF02981942. PMID: 11372736.
  12. Fu RT, Xue HM, Xu HG, Huang K, Fang JP, Huang SL, et al. [Therapeutic efficacy evaluation of rabbit anti-thymocyte globulin combined with cyclosporine A in children with aplastic anemia]. Zhongguo Shi Yan Xue Ye Xue Za Zhi. 2013; 21: 426-430. Chinese. doi: 10.7534/j. issn.1009-2137.2013.02.035. PMID: 23628047.
  13. Mariathasan S, Turley SJ, Nickles D, Castiglioni A, Yuen K, Wang Y, et al. TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature. 2018; 554: 544-548. doi: 10.1038/nature25501. Epub 2018 Feb 14. PMID: 29443960; PMCID: PMC6028240.
  14. Tsukamoto H, Fujieda K, Miyashita A, Fukushima S, Ikeda T, Kubo Y, et al. Combined Blockade of IL6 and PD-1/PD-L1 Signaling Abrogates Mutual Regulation of Their Immunosuppressive Effects in the Tumor Microenvironment. Cancer Res. 2018; 78: 5011-5022. doi: 10.1158/0008-5472.CAN-18-0118. Epub 2018 Jul 2. PMID: 29967259.
  15. Yonekura K, Takeda K, Kawakami N, Kanzaki T, Kanekura T, Utsunomiya A. Therapeutic Efficacy of Etretinate on Cutaneous-type Adult T-cell Leukemia-Lymphoma. Acta Derm Venereol. 2019; 99: 774-776. doi: 10.2340/00015555-3196. PMID: 31017247.
  16. Zhu F, Qiao J, Zhong XM, Wu QY, Chen W, Yao Y, et al. Antithymocyte globulin combined with cyclosporine A down-regulates T helper 1 cells by modulating T cell immune response cDNA 7 in aplastic anemia. Med Oncol. 2015; 32: 197. doi: 10.1007/s12032-015-0647-2. Epub 2015 Jun 7. PMID: 26049920.
  17. Toofan P, Wheadon H. Role of the bone morphogenic protein pathway in developmental haemopoiesis and leukaemogenesis. Biochem Soc Trans. 2016; 44: 1455-1463. doi: 10.1042/BST20160104. PMID:27911727.
  18. Gordon-Smith, T. Haemopoiesis-the formation of blood cells. Medicine. 2013; 41: 200-203.
  19. Hamzic E, Whiting K, Gordon Smith E, Pettengell R. Characterization of bone marrow mesenchymal stromal cells in aplastic anaemia. Br J Haematol. 2015; 169: 804-813. doi: 10.1111/bjh.13364. Epub 2015 Mar 29. PMID: 25819548.
  20. Marsh JC, Chang J, Testa NG, Hows JM, Dexter TM. In vitro assessment of marrow ‘stem cell’ and stromal cell function in aplastic anaemia. Br J Haematol. 1991; 78: 258-267. doi: 10.1111/j.1365-2141.1991. tb04426.x. PMID: 1712224.
  21. Tripathy NK, Singh SP, Nityanand S. Enhanced adipogenicity of bone marrow mesenchymal stem cells in aplastic anemia. Stem Cells Int. 2014; 2014: 276862. doi: 10.1155/2014/276862. Epub 2014 Apr 30. PMID: 24876847; PMCID: PMC4021843.
  22. Qu Y, Lin Q, Yuan Y, Sun Z, Li P, Wang F, et al. Cyclosporin A inhibits adipogenic differentiation and regulates immunomodulatory functions of murine mesenchymal stem cells. Biochem Biophys Res Commun. 2018; 498: 516-522. doi: 10.1016/j.bbrc.2018.03.012. Epub 2018 Mar 3. PMID: 29510137.
  23. Song LH, Pan W, Yu YH, Quarles LD, Zhou HH, Xiao ZS. Resveratrol prevents CsA inhibition of proliferation and osteoblastic differentiation of mouse bone marrow-derived mesenchymal stem cells through an ER/NO/cGMP pathway. Toxicol In Vitro. 2006; 20: 915-922. doi: 10.1016/j.tiv.2006.01.016. Epub 2006 Mar 9. PMID: 16524694.
  24. Rajesh KG, Sasaguri S, Zhitian Z, Suzuki R, Asakai R, Maeda H. Second window of ischemic preconditioning regulates mitochondrial permeability transition pore by enhancing Bcl-2 expression. Cardiovasc  Res.  2003;  59:  297-307.  doi:  10.1016/s0008-6363(03)00358-4. PMID: 12909313.
  25. Tan C, Dlugosz PJ, Peng J, Zhang Z, Lapolla SM, Plafker SM,et al. Auto- activation of the apoptosis protein Bax increases mitochondrial membrane permeability and is inhibited by Bcl-2. J Biol Chem. 2006; 281: 14764-14775. doi: 10.1074/jbc.M602374200. Epub 2006 Mar 29. PMID: 16571718; PMCID: PMC2826894.
  26. Li J, Yang S, Lu S, Zhao H, Feng J, Li W, et al, Zhang L, Zheng Y, Han ZC. Differential gene expression profile associated with the abnormality of bone marrow mesenchymal stem cells in aplastic anemia. PLoS One. 2012; 7: e47764. doi: 10.1371/journal.pone.0047764. Epub 2012 Nov 5. PMID: 23144828; PMCID: PMC3489901.
  27. Bacigalupo A, Valle M, Podestà M, Pitto A, Zocchi E, De Flora A, et al. T-cell suppression mediated by mesenchymal stem cells is deficient in patients with severe aplastic anemia. Exp Hematol. 2005; 33: 819- 827. doi: 10.1016/j.exphem.2005.05.006. PMID: 15963858.
  28. Rizzo S, Killick SB, Patel S, Ball SE, Wadhwa M, Dilger P, et al. Reduced TGF-beta1 in patients with aplastic anaemia in vivo and in vitro. Br J Haematol. 1999; 107: 797-803. doi: 10.1046/j.1365- 2141.1999.01761.x. PMID: 10606887.
  29. Taketazu F, Miyagawa K, Ichijo H, Oshimi K, Mizoguchi H, Hirai H, et al. Decreased level of transforming growth factor-beta in blood lymphocytes of patients with aplastic anemia. Growth Factors. 1992; 6: 85-90. doi: 10.3109/08977199209008874. PMID: 1591018.
  30. Tsurutani Y, Fujimoto M, Takemoto M, Irisuna H, Koshizaka M, Onishi S, et al. The roles of transforming growth factor-β and Smad3 signaling in adipocyte differentiation and obesity. Biochem Biophys Res Commun. 2011; 407: 68-73. doi: 10.1016/j.bbrc.2011.02.106. Epub 2011 Feb 26. PMID: 21356196.
  31. Mu Y, Gudey SK, Landström M. Non-Smad signaling pathways. Cell Tissue Res. 2012; 347: 11-20. doi: 10.1007/s00441-011-1201-y. Epub 2011 Jun 24. PMID: 21701805.
  32. Pedrosa M, Gomes J, Laranjeira P, Duarte C, Pedreiro S, Antunes B, et al. Immunomodulatory effect of human bone marrow-derived mesenchymal stromal/stem cells on peripheral blood T cells from rheumatoid arthritis patients. J Tissue Eng Regen Med. 2020; 14: 16-28. doi: 10.1002/term.2958. Epub 2019 Nov 11. PMID: 31502378.
  33. Liu M, Zeng X, Wang J, Fu Z, Wang J, Liu M, et al. Immunomodulation by mesenchymal stem cells in treating human autoimmune disease- associated lung fibrosis. Stem Cell Res Ther. 2016; 7: 63. doi: 10.1186/ s13287-016-0319-y. PMID: 27107963; PMCID: PMC4842299.
  34. Zheng GP, Ge MH, Shu Q, Rojas M, Xu J. Mesenchymal stem cells in the treatment of pediatric diseases. World J Pediatr. 2013; 9: 197- 211. doi: 10.1007/s12519-013-0425-1. Epub 2013 Aug 9. PMID: 23929252.
  35. Gupta V, Kumar S, Sonowal R, Singh SK. Interleukin-6 and Interleukin-8 Levels Correlate With the Severity of Aplastic Anemia in Children. J Pediatr Hematol Oncol. 2017; 39: 214-216. doi: 10.1097/ MPH.0000000000000724. PMID: 28060106.
  36. Schrezenmeier H, Marsh JC, Stromeyer P, Müller H, Heimpel H, Gordon-Smith EC, et al. A phase I/II trial of recombinant human interleukin-6 in patients with aplastic anaemia. Br J Haematol. 1995; 90: 283-292. doi: 10.1111/j.1365-2141.1995.tb05148.x. PMID:7794747.
  37. Lu S, Qiao X, Xie X. Elevated Serum Interleukin-6 Predicts Favorable Response to Immunosuppressive Therapy in Children With Aplastic Anemia. J Pediatr Hematol Oncol. 2017; 39: 614-617. doi: 10.1097/ MPH.0000000000000942. PMID: 29068868.
  38. Okazaki T, Wang J. PD-1/PD-L pathway and autoimmunity. Autoimmunity. 2005; 38: 353-357. doi: 10.1080/08916930500124072. PMID: 16227150.
  39. Francisco LM, Sage PT, Sharpe AH. The PD-1 pathway in tolerance and autoimmunity. Immunol Rev. 2010; 236: 219-242. doi: 10.1111/j.1600-065X.2010.00923.x. PMID: 20636820; PMCID: PMC2919275.
  40. Gianchecchi E, Delfino DV, Fierabracci A. Recent insights into the role of the PD-1/PD-L1 pathway in immunological tolerance and autoimmunity. Autoimmun Rev. 2013; 12: 1091-1100. doi: 10.1016/j.autrev.2013.05.003. Epub 2013 Jun 20. PMID: 23792703.
  41. Michot JM, Vargaftig J, Leduc C, Quere G, Burroni B, Lazarovici J, et al. Immune-related bone marrow failure following anti-PD1 therapy. Eur J Cancer. 2017; 80: 1-4. doi: 10.1016/j.ejca.2017.04.004. Epub 2017 May 17. PMID: 28527392.
  42. David JM, Dominguez C, McCampbell KK, Gulley JL, Schlom J, Palena C. A novel bifunctional anti-PD-L1/TGF-β Trap fusion protein (M7824) efficiently reverts mesenchymalization of human lung cancer cells. Oncoimmunology. 2017; 6: e1349589. doi: 10.1080/2162402X.2017.1349589. PMID: 29123964; PMCID: PMC5665067.

Shi LF, Sun L, Ma BD, Guo ZQ, Jin RR, et al. (2024) Cyclosporin a Inhibits Adipogenic Differentiation through the TGF- β1 Signaling Pathway and Regulates Immunomodulatory Functions of Mesenchymal Stem Cells in Aplastic Anemia Patients. Arch Stem Cell Res 6(1): 1020

Received : 10 May 2024
Accepted : 14 Jun 2024
Published : 17 Jun 2024
Journals
Annals of Otolaryngology and Rhinology
ISSN : 2379-948X
Launched : 2014
JSM Schizophrenia
Launched : 2016
Journal of Nausea
Launched : 2020
JSM Internal Medicine
Launched : 2016
JSM Hepatitis
Launched : 2016
JSM Oro Facial Surgeries
ISSN : 2578-3211
Launched : 2016
Journal of Human Nutrition and Food Science
ISSN : 2333-6706
Launched : 2013
JSM Regenerative Medicine and Bioengineering
ISSN : 2379-0490
Launched : 2013
JSM Spine
ISSN : 2578-3181
Launched : 2016
Archives of Palliative Care
ISSN : 2573-1165
Launched : 2016
JSM Nutritional Disorders
ISSN : 2578-3203
Launched : 2017
Annals of Neurodegenerative Disorders
ISSN : 2476-2032
Launched : 2016
Journal of Fever
ISSN : 2641-7782
Launched : 2017
JSM Bone Marrow Research
ISSN : 2578-3351
Launched : 2016
JSM Mathematics and Statistics
ISSN : 2578-3173
Launched : 2014
Journal of Autoimmunity and Research
ISSN : 2573-1173
Launched : 2014
JSM Arthritis
ISSN : 2475-9155
Launched : 2016
JSM Head and Neck Cancer-Cases and Reviews
ISSN : 2573-1610
Launched : 2016
JSM General Surgery Cases and Images
ISSN : 2573-1564
Launched : 2016
JSM Anatomy and Physiology
ISSN : 2573-1262
Launched : 2016
JSM Dental Surgery
ISSN : 2573-1548
Launched : 2016
Annals of Emergency Surgery
ISSN : 2573-1017
Launched : 2016
Annals of Mens Health and Wellness
ISSN : 2641-7707
Launched : 2017
Journal of Preventive Medicine and Health Care
ISSN : 2576-0084
Launched : 2018
Journal of Chronic Diseases and Management
ISSN : 2573-1300
Launched : 2016
Annals of Vaccines and Immunization
ISSN : 2378-9379
Launched : 2014
JSM Heart Surgery Cases and Images
ISSN : 2578-3157
Launched : 2016
Annals of Reproductive Medicine and Treatment
ISSN : 2573-1092
Launched : 2016
JSM Brain Science
ISSN : 2573-1289
Launched : 2016
JSM Biomarkers
ISSN : 2578-3815
Launched : 2014
JSM Biology
ISSN : 2475-9392
Launched : 2016
Annals of Clinical and Medical Microbiology
ISSN : 2578-3629
Launched : 2014
JSM Pediatric Surgery
ISSN : 2578-3149
Launched : 2017
Journal of Memory Disorder and Rehabilitation
ISSN : 2578-319X
Launched : 2016
JSM Tropical Medicine and Research
ISSN : 2578-3165
Launched : 2016
JSM Head and Face Medicine
ISSN : 2578-3793
Launched : 2016
JSM Cardiothoracic Surgery
ISSN : 2573-1297
Launched : 2016
JSM Bone and Joint Diseases
ISSN : 2578-3351
Launched : 2017
JSM Bioavailability and Bioequivalence
ISSN : 2641-7812
Launched : 2017
JSM Atherosclerosis
ISSN : 2573-1270
Launched : 2016
Journal of Genitourinary Disorders
ISSN : 2641-7790
Launched : 2017
Journal of Fractures and Sprains
ISSN : 2578-3831
Launched : 2016
Journal of Autism and Epilepsy
ISSN : 2641-7774
Launched : 2016
Annals of Marine Biology and Research
ISSN : 2573-105X
Launched : 2014
JSM Health Education & Primary Health Care
ISSN : 2578-3777
Launched : 2016
JSM Communication Disorders
ISSN : 2578-3807
Launched : 2016
Annals of Musculoskeletal Disorders
ISSN : 2578-3599
Launched : 2016
Annals of Virology and Research
ISSN : 2573-1122
Launched : 2014
JSM Renal Medicine
ISSN : 2573-1637
Launched : 2016
Journal of Muscle Health
ISSN : 2578-3823
Launched : 2016
JSM Genetics and Genomics
ISSN : 2334-1823
Launched : 2013
JSM Anxiety and Depression
ISSN : 2475-9139
Launched : 2016
Clinical Journal of Heart Diseases
ISSN : 2641-7766
Launched : 2016
Annals of Medicinal Chemistry and Research
ISSN : 2378-9336
Launched : 2014
JSM Pain and Management
ISSN : 2578-3378
Launched : 2016
JSM Women's Health
ISSN : 2578-3696
Launched : 2016
Clinical Research in HIV or AIDS
ISSN : 2374-0094
Launched : 2013
Journal of Endocrinology, Diabetes and Obesity
ISSN : 2333-6692
Launched : 2013
Journal of Substance Abuse and Alcoholism
ISSN : 2373-9363
Launched : 2013
JSM Neurosurgery and Spine
ISSN : 2373-9479
Launched : 2013
Journal of Liver and Clinical Research
ISSN : 2379-0830
Launched : 2014
Journal of Drug Design and Research
ISSN : 2379-089X
Launched : 2014
JSM Clinical Oncology and Research
ISSN : 2373-938X
Launched : 2013
JSM Bioinformatics, Genomics and Proteomics
ISSN : 2576-1102
Launched : 2014
JSM Chemistry
ISSN : 2334-1831
Launched : 2013
Journal of Trauma and Care
ISSN : 2573-1246
Launched : 2014
JSM Surgical Oncology and Research
ISSN : 2578-3688
Launched : 2016
Annals of Food Processing and Preservation
ISSN : 2573-1033
Launched : 2016
Journal of Radiology and Radiation Therapy
ISSN : 2333-7095
Launched : 2013
JSM Physical Medicine and Rehabilitation
ISSN : 2578-3572
Launched : 2016
Annals of Clinical Pathology
ISSN : 2373-9282
Launched : 2013
Annals of Cardiovascular Diseases
ISSN : 2641-7731
Launched : 2016
Journal of Behavior
ISSN : 2576-0076
Launched : 2016
Annals of Clinical and Experimental Metabolism
ISSN : 2572-2492
Launched : 2016
Clinical Research in Infectious Diseases
ISSN : 2379-0636
Launched : 2013
JSM Microbiology
ISSN : 2333-6455
Launched : 2013
Journal of Urology and Research
ISSN : 2379-951X
Launched : 2014
Journal of Family Medicine and Community Health
ISSN : 2379-0547
Launched : 2013
Annals of Pregnancy and Care
ISSN : 2578-336X
Launched : 2017
JSM Cell and Developmental Biology
ISSN : 2379-061X
Launched : 2013
Annals of Aquaculture and Research
ISSN : 2379-0881
Launched : 2014
Clinical Research in Pulmonology
ISSN : 2333-6625
Launched : 2013
Journal of Immunology and Clinical Research
ISSN : 2333-6714
Launched : 2013
Annals of Forensic Research and Analysis
ISSN : 2378-9476
Launched : 2014
JSM Biochemistry and Molecular Biology
ISSN : 2333-7109
Launched : 2013
Annals of Breast Cancer Research
ISSN : 2641-7685
Launched : 2016
Annals of Gerontology and Geriatric Research
ISSN : 2378-9409
Launched : 2014
Journal of Sleep Medicine and Disorders
ISSN : 2379-0822
Launched : 2014
JSM Burns and Trauma
ISSN : 2475-9406
Launched : 2016
Chemical Engineering and Process Techniques
ISSN : 2333-6633
Launched : 2013
Annals of Clinical Cytology and Pathology
ISSN : 2475-9430
Launched : 2014
JSM Allergy and Asthma
ISSN : 2573-1254
Launched : 2016
Journal of Neurological Disorders and Stroke
ISSN : 2334-2307
Launched : 2013
Annals of Sports Medicine and Research
ISSN : 2379-0571
Launched : 2014
JSM Sexual Medicine
ISSN : 2578-3718
Launched : 2016
Annals of Vascular Medicine and Research
ISSN : 2378-9344
Launched : 2014
JSM Biotechnology and Biomedical Engineering
ISSN : 2333-7117
Launched : 2013
Journal of Hematology and Transfusion
ISSN : 2333-6684
Launched : 2013
JSM Environmental Science and Ecology
ISSN : 2333-7141
Launched : 2013
Journal of Cardiology and Clinical Research
ISSN : 2333-6676
Launched : 2013
JSM Nanotechnology and Nanomedicine
ISSN : 2334-1815
Launched : 2013
Journal of Ear, Nose and Throat Disorders
ISSN : 2475-9473
Launched : 2016
JSM Ophthalmology
ISSN : 2333-6447
Launched : 2013
Journal of Pharmacology and Clinical Toxicology
ISSN : 2333-7079
Launched : 2013
Annals of Psychiatry and Mental Health
ISSN : 2374-0124
Launched : 2013
Medical Journal of Obstetrics and Gynecology
ISSN : 2333-6439
Launched : 2013
Annals of Pediatrics and Child Health
ISSN : 2373-9312
Launched : 2013
JSM Clinical Pharmaceutics
ISSN : 2379-9498
Launched : 2014
JSM Foot and Ankle
ISSN : 2475-9112
Launched : 2016
JSM Alzheimer's Disease and Related Dementia
ISSN : 2378-9565
Launched : 2014
Journal of Addiction Medicine and Therapy
ISSN : 2333-665X
Launched : 2013
Journal of Veterinary Medicine and Research
ISSN : 2378-931X
Launched : 2013
Annals of Public Health and Research
ISSN : 2378-9328
Launched : 2014
Annals of Orthopedics and Rheumatology
ISSN : 2373-9290
Launched : 2013
Journal of Clinical Nephrology and Research
ISSN : 2379-0652
Launched : 2014
Annals of Community Medicine and Practice
ISSN : 2475-9465
Launched : 2014
Annals of Biometrics and Biostatistics
ISSN : 2374-0116
Launched : 2013
JSM Clinical Case Reports
ISSN : 2373-9819
Launched : 2013
Journal of Cancer Biology and Research
ISSN : 2373-9436
Launched : 2013
Journal of Surgery and Transplantation Science
ISSN : 2379-0911
Launched : 2013
Journal of Dermatology and Clinical Research
ISSN : 2373-9371
Launched : 2013
JSM Gastroenterology and Hepatology
ISSN : 2373-9487
Launched : 2013
Annals of Nursing and Practice
ISSN : 2379-9501
Launched : 2014
JSM Dentistry
ISSN : 2333-7133
Launched : 2013
Author Information X