Human Umbilical Cord Mesenchymal Stem Cells in Diabetes Mellitus and Its Complications: Applications and Research Advances
Human Umbilical Cord Mesenchymal Stem Cells in Diabetes Mellitus and Its Complications: Applications and Research Advances
Human Umbilical Cord Mesenchymal Stem Cells in Diabetes Mellitus and Its Complications: Applications and Research Advances
20 1492
Ivyspring
International Publisher International Journal of Medical Sciences
2023; 20(11): 1492-1507. doi: 10.7150/ijms.87472
Review
Corresponding author: Department of Endocrinology, the Second Hospital of Jilin University, Changchun 130041, Jilin, P.R. China; E-mail:
wangs93@sina.com.
© The author(s). This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/).
See http://ivyspring.com/terms for full terms and conditions.
Abstract
Diabetes mellitus and its complications pose a major threat to global health and affect the quality of life
and life expectancy of patients. Currently, the application of traditional therapeutic drugs for diabetes
mellitus has great limitations and can only temporarily control blood glucose but not fundamentally cure
it. Mesenchymal stem cells, as pluripotent stromal cells, have multidirectional differentiation potential,
high self-renewal, immune regulation, and low immunogenicity, which provide a new idea and possible
development direction for diabetes mellitus treatment. Regenerative medicine with mesenchymal stem
cells treatment as the core treatment will become another treatment option for diabetes mellitus after
traditional treatment. Recently, human umbilical cord mesenchymal stem cells have been widely used in
basic and clinical research on diabetes mellitus and its complications because of their abundance, low
ethical controversy, low risk of infection, and high proliferation and differentiation ability. This paper
reviews the therapeutic role and mechanism of human umbilical cord mesenchymal stem cells in diabetes
mellitus and its complications and highlights the challenges faced by the clinical application of human
umbilical cord mesenchymal stem cells to provide a more theoretical basis for the application of human
umbilical cord mesenchymal stem cells in diabetes mellitus patients.
Keywords: umbilical cord, mesenchymal stem cells, diabetes mellitus, pluripotent, clinical application, regenerative medicine
Introduction
Diabetes mellitus (DM) is the most prevalent with a combination of impaired hypoglycaemic
metabolic disorder caused by the inability of the awareness and severe hypoglycaemic episodes (2).
pancreas to secrete insulin adequately or the body’s However, islet transplantation may be greatly limited
inability to use insulin effectively. According to the in clinical application due to a shortage of donor islets
9th edition of the International Diabetes Federation and immune rejection. Drug therapy is an important
(IDF) Atlas of Diabetes, approximately 7.002 million treatment modality for patients with T2DM (3), but its
adults aged 20–79 years will have DM worldwide by side effects (such as diarrhoea, nausea, vomiting, and
2045 (1). Type 1 diabetes mellitus (TIDM) and type 2 anaemia) and drug prices remain to be investigated.
diabetes mellitus (T2DM) are the two most common Meanwhile, persistent hyperglycaemia can cause
types of DM. Patients with T1DM are primarily chronic damage or dysfunction of the eyes, kidneys,
treated with insulin replacement therapy to alleviate heart, blood vessels, and nerves, and intervention in
absolute insulin deficiency, but may be at risk for DM and its complications and reduction of mortality
hypoglycaemia and tumourigenesis. Human islet are imminent (4, 5).
transplantation is an effective treatment for T1DM, Mesenchymal stem cells (MSCs) are widely used
https://www.medsci.org
Int. J. Med. Sci. 2023, Vol. 20 1493
in various cell therapies because of their many potential and proliferative capacity (18). Compared
advantages, such as self-renewal capacity, multi- with dental pulp-derived mesenchymal stem cells
spectral differentiation ability, tissue damage repair, (PU-MSCs) and adipose tissue-derived mesenchymal
and lack of co-stimulatory molecules (6). The stem cells (AD-MSCs), HUC-MSCs have the strongest
abundant source of human umbilical cord efficacy in ameliorating glucose and lipid metabolism
mesenchymal stem cells (HUC-MSCs), low ethical disorders in T2DM (19). (8) Very low immunogenicity
controversy, low infection risk, high proliferation and (20). In summary, HUC-MSCs are an ideal source of
differentiation ability, and very low immunogenicity cells for cell therapy in DM.
make them uniquely advantageous for DM therapy.
Recently, studies related to the treatment of DM with Possible mechanisms of HUC-MSCs for
HUC-MSCs have rapidly developed. This review DM treatment
describes the advantages and mechanisms of MSCs for DM are cell-based therapeutic
HUC-MSCs in treating DM and the application and approaches that have shown remarkable therapeutic
research progress of HUC-MSCs in DM-related effects in DM because of their self-renewal,
complications, providing more options for managing differentiation potential, and immunosuppressive
DM and its complications. properties. Numerous studies have shown that
HUC-MSCs are a novel strategy to treat DM, and their
Source of HUC-MSCs possible mechanisms (21) include: 1) homing to the
Umbilical cord blood (UCB) is a valuable stem damaged pancreas and acting through local nutrition
cell source. MSCs can be isolated from neonatal and secretion of paracrine factors; 2) differentiation
umbilical cords by enzymatic digestion and show a into insulin-producing cells (IPCs); 3) reversal of
positive expression of classical MSC surface markers. beta-cell (β-cell) dedifferentiation, thereby alleviating
The umbilical cord comprises the umbilical artery, β-cell dysfunction and protecting islet β-cells; 4)
umbilical vein, Wharton's jelly (WJ), and external promotion of islet β-cell regeneration; 5) secretion of
amniotic epithelium surrounding the mucus anti-inflammatory cytokines and macrophage
connective tissue (7). MSCs can be isolated from phenotype regulation, thereby reducing islet β-cell
different umbilical cord parts, including the blood, inflammation; and 6) enhancing insulin sensitivity in
sub-umbilical vein endothelium, and the WJ. target tissues and improving insulin resistance (Fig.
Researchers have successfully isolated and cultured 1).
MSCs from the perivascular layer of Wharton
collagenous vessels of the human umbilical vein (8). Homing effect of HUC-MSCs
MSCs can also be isolated from non-perivascular One advantage of MSCs for DM mitigation is
areas (sub-amniotic membranes) (9). Platelet-derived their ability to home to damaged tissues and then
growth factor (PDGF) produced by human amniotic directly proliferate and differentiate to replace
cells may induce cell migration from the vascular damaged cells and repair damaged tissues. Homing is
system to the amnion (10). The human umbilical cord potentially important for recruiting MSCs to the
is a rich MSC source. injury and regeneration sites (22). MSCs homing
includes both non-systematic and systemic homing.
Advantages of HUC-MSCs In non-systematic homing, MSCs are locally
HUC-MSCs have compelling advantages in transplanted into the target tissue and then directed to
treating DM, including (1) abundant sources, easy the injury site via a chemokine gradient. In systemic
collection, and easy preservation and transportation homing, the molecular mechanisms of MSCs homing
(11); (2) easy isolation, high purity, and include initial tethering by selectins, activation by
non-tumorigenic (12); (3) high amplification potential cytokines, blockade by integrins, exudation or
(13); (4) functional stability after lyophilisation and migration using matrix remodelling agents, and
recovery (14); (5) no adverse effects of collection on extravasation toward chemokine gradients (23). In
the donor, and ethical issues are circumvented (15); 2017, HUC-MSCs labelled with 1,1'-dioctadecyl-
and (6) low probability of infection and transmission 3,3,3',3'-tetramethylindocarbocyanine perchlorate
of pathogenic microorganisms. In contrast, bone (DiI) were detected in the pancreas of T1DM mice,
marrow-derived MSCs (BM-MSCs) have a high risk of suggesting that HUC-MSCs may target and migrate to
viral infection and a significant decrease in cell damaged organs to exert therapeutic effects (24). Yin
number and proliferation/differentiation capacity et al. pre-labelled HUC-MSCs with cell membrane-Dil
with age (16, 17). (7) More primitive and proliferative (CM-Dil) to demonstrate their migration in various
differentiation capacity. Compared with BM-MSCs, tissues, thus confirming the implantation of MSCs in
HUC-MSCs have higher pancreatic differentiation the pancreatic islets of T2DM mice. This suggests that
https://www.medsci.org
Int. J. Med. Sci. 2023, Vol. 20 1494
homing of HUC-MSCs may be closely related to tissue healing, wound healing, modulating immunity,
damage (25). Overall, HUC-MSCs may play a role in anti-inflammation, anti-apoptosis, and cytoprotection.
treating DM by homing to damaged islets. However, Recently, there has been intense interest in the
the homing rate of MSCs is low, and MSCs may exert synthesis and release of EVs by MSCs via paracrine
protective effects through other mechanisms. secretion. Human umbilical cord mesenchymal stem
cell-derived exosomes (HucMSC-exs) are nanometer-
Paracrine effects of HUC-MSCs sized and are capable of rapid diffusion across
The paracrine properties of MSCs make them a biological barriers and cell membranes. Numerous
key tissue repair option, and the paracrine effect of studies have shown that HucMSC-exs have
MSCs is achieved through the secretion of soluble anti-inflammatory, anti-apoptotic, tissue repair,
factors and release of extracellular vehicles (EVs), neuroprotective, and immunomodulatory properties,
such as exosomes and microvesicles (26). All the suggesting that HucMSC-ex may be a potential DM
factors secreted by MSCs are called the secretome and therapy. HucMSC-ex alleviates T2DM by activating
comprise various cytokines, chemokines, angiogenic the regenerative capacity of islets (31), improving
factors, and growth factors. Moreover, up to 80% of insulin sensitivity (32), reversing peripheral insulin
the therapeutic effects of MSCs are mediated by resistance, and attenuating β-cell destruction (33).
paracrine signalling (27). HUC-MSCs secrete soluble Human umbilical cord mesenchymal stem cell-
molecules such as keratinocyte growth factor (KGF), derived small extracellular vesicles (HUC-MSC-sEVs)
hepatocyte growth factor (HGF), vascular endothelial attenuated structural damage in the pancreas, kidney,
growth factor (VEGF), fibroblast growth factor (FGF), and liver of T2DM rats (34). HucMSC-ex protects
placental growth factor (PGF), monocyte chemo- β-cells from hypoxia-induced apoptosis by carrying
attractant protein 1 (MCP-1), insulin-like growth miR-21 to attenuate endoplasmic reticulum (ER)
factor 1 (IGF-1), epidermal growth factor (EGF), stress and inhibit p38 MAPK phosphorylation (35).
prostaglandin E2 (PGE2), indoleamine 2,3-deoxy- Exosome-loaded immunomodulatory biomaterials
genase (IDO), interleukin-10 (IL-10), interleukin-6 can attenuate the local immune response induced by
(IL-6), transforming growth factor-β1 (TGF-β1), nitric grafts in DM mice (36). The above studies revealed the
oxide (NO), human leukocyte antigen-G5 (HLA-G5), potential value of HucMSC-ex and miRs in DM.
tumour necrosis factor-α stimulated gene 6 (TSG-6), Overall, HUC-MSCs play a protective role against DM
and neurotrophic factors (12, 28-30). These factors by secreting soluble factors and EVs.
play a role in promoting tissue regeneration,
participating in angiogenesis, promoting ulcer tissue
Figure 1. This figure illustrates the possible mechanism of HUC-MSCs for the treatment of DM. HUC-MSCs exert beneficial effects on diabetes by differentiating into IPCs,
promoting islet β-cell regeneration, reversing β-cell dedifferentiation, reducing islet β-cell inflammation, and improving insulin resistance. IPCs insulin-producing cells.
https://www.medsci.org
Int. J. Med. Sci. 2023, Vol. 20 1495
Differentiation of HUC-MSCs into IPCs tiation into functionally competent β-cells. Pax4, in
concert with Pdx1, Ngn3, and MAF bZIP transcription
HUC-MSCs ameliorate hyperglycaemia and
factor A (MafA), can induce the differentiation of
weight loss in DM rats by differentiating them into
HUC-MSCs into pancreatic β-like cells (pβLCs)
IPCs (37-39). HUC-MSCs induced to differentiate into
functional pancreatic β cells (63). MSCs participate in
IPCs (40, 41) express pancreatic β-cell differentiation-
the repair process by secreting various cytokines and
related genes (e.g. nestin, pancreatic duodenal
growth factors with paracrine and autocrine activities,
homeobox-1 (PDX-1), neurogenin3 (NGN3), paired
which may contribute to endogenous β-cell
box 6 (PAX6), paired box 4 (PAX4), nk2 homeobox 2
regeneration and islet structural recovery (21). Wei et
(NKX2.2), nk6 homeobox 1 (NKX6.1), glucose
al. found that HUC-MSCs protect islets from
transporter 2 (GLUT-2), and insulin (INS) genes) (42,
hypoxia-induced dysfunction (64) and secrete IGF-1
43), and promote the secretion of serum C-peptide
to exert a trophic effect on islets (65). Bao et al. found
and INS in DM rats (44, 45). Additionally, HUC-MSCs
that HUC-MSCs overexpressing tissue inhibitors of
promote the survival, function, and number of
matrix metalloproteinase (TIMP)-1 induced weight
islet-like cell clusters (46). Co-culture of HUC-MSCs
loss and hypoglycaemia and improved islet function
with T1DM rat pancreatic cells promotes survival,
and survival in T1DM mice (66). Lu et al. found that
proliferation, and induced differentiation of HUC-
HUC-MSC transplantation is safe and effective in
MSCs into IPCs (47). Furthermore, the differentiation
T1DM patients and may better protect residual β-cells
of IPCs is a very complex process, and the initial stage
(67). Hu et al. found that the combination of
of nestin preselection, appropriate induction reagents
HUC-MSCs and selegiline was effective in improving
(48), and extracellular matrix (49) are necessary for the
hyperglycaemia, promoting islet β-cell regeneration,
in vitro culture of IPCs from HUC-MSCs. PDX-1 (50,
and inhibiting islet alpha cell (α-cell) production in
51), inhibition of Notch signalling (52) and laminin
T2DM rats (68). Although the exact mechanism needs
411 (53) effectively regulate the differentiation of
to be further explored, this study may provide a new
MSCs into IPCs. Under hypoxic conditions,
therapeutic approach for DM.
UCB-MSCs also efficiently differentiate into IPCs (54,
55). Additionally, factors that effectively promote the Interaction of HUC-MSCs with various
efficacy of IPC action include Port-A catheter immune cells and cytokines
transplantation (56), suspension culture (57), and
addition of the histone deacetylase (HDAC) inhibitor HUC-MSCs and macrophage polarization
TMP269 (58). Overall, HUC-MSCs can replace DM is characterised by mild chronic inflam-
damaged islet β cells by inducing differentiation into mation, which is often accompanied by inflammatory
IPCs, which are ideal seed cells to treat DM. cell infiltration in islets. Macrophage infiltration of
islets and autoimmune destruction of β-cells are
HUC-MSCs can effectively improve islet β-cell important features of the chronic inflammatory
function process in T1DM. Macrophages may be a major
β-Cell dedifferentiation is thought to be an contributor to the development of chronic
important contributor to β-cell dysfunction in T2DM inflammation and insulin resistance in patients with
(59). Pro-inflammatory cytokines can lead to β-cell T2DM (3). UC-MSC transplantation induces an
dysfunction and de-differentiation. MSCs reduce increase in M2 macrophages in pancreatic islets,
endogenous interleukin-1b (IL-1b) production in adipose tissue, liver, and skeletal muscle. HUC-MSCs
T2DM islets by secreting IL-1Ra, thereby reducing produce anti-inflammatory mediators and growth
islet injury and reversing β-cell dedifferentiation (60). factors that suppress inflammation and improve
Additionally, it has been shown that the interleukin-1 insulin sensitivity and β-cell regeneration (25).
receptor antagonist (IL-1Ra) can also regulate the HUC-MSCs reduce insulin resistance by secreting IL-6
phenotypic transition of macrophages (61). In db/db (69) and IL-10 (70) to promote M2 macrophage
mice, early infusion of HUC-MSCs reduced β-cell polarization. MCP-1 secreted by HUC-MSCs
dedifferentiation markers, such as aldehyde synergistically regulates macrophage polarisation
dehydrogenase 1 family member A3+ (ALDH1A3+), with IL-6 (71). Additionally, low-dose decitabine may
and increased the proportion of Ins+ β cells and prolong the antidiabetic effects of MSCs and promote
Pdx1+/Ins+ cells (62). This suggests that MSCs sustainable β-cell recovery by polarising macrophages
transplantation may be a therapeutic strategy for to the M2 phenotype (72). Overall, HUC-MSCs can
protecting and restoring β-cell function in patients reduce islet β-cell inflammation by polarising
with T2DM. Additionally, the potential mechanisms macrophages to the M2 anti-inflammatory phenotype,
for the therapeutic effects of MSCs on DM may thereby alleviating islet dysfunction in patients with
involve islet regeneration, including direct differen- DM.
https://www.medsci.org
Int. J. Med. Sci. 2023, Vol. 20 1496
HUC-MSCs and other immune cells conclusion, HUC-MSCs act as an effective treatment
MSCs not only act on innate immune cells but for T2DM by improving IR, thereby providing a
also interact with other immune cells, thus regulating potential avenue for developing novel clinical T2DM
multiple effector functions (73). MSCs regulate therapies.
antigen presentation by dendritic cells (DCs),
cytotoxicity of natural killer (NK) cells and neutrophil
HUC-MSCs and other types of diabetes
activation. MSCs induce peripheral tolerance in T cells Recently, with advances in regenerative
and exert effective tissue protection through the medicine research, HUC-MSCs may provide a new
release of anti-inflammatory, anti-apoptotic, and treatment option for other types of DM. Hu et al.
trophic molecules (74). Li et al. found that regulatory found that HUC-MSCs therapy could restore the
T cells (Treg)/T helper cell 17 (Th17) and Treg/T function of residual islet β cells in patients with
helper cell 1 (Th1) cell ratios increased significantly new-onset T1DM over a longer period. This suggests
after 4 weeks of transplantation of HUC-MSCs, while that implantation of HUC-MSCs is expected to be an
the Th17/Th1 cell ratio remained unchanged (75), effective strategy for treating new-onset T1DM (87).
suggesting that HUC-MSCs ameliorate immune Yang et al. found that HUC-MSCs reduced
disorders in T2DM by repairing Treg cells. HUC- inflammatory responses and attenuated pancreatic
MSCs can reduce blood glucose, increase C-peptide injury in rats with severe acute pancreatitis (SAP) (88).
levels, and Treg production in T2DM patients (76), Kong et al. found that HUC-MSCs ameliorated
suggesting that HUC-MSCs with powerful chronic pancreatitis in rats via the AKT-mTOR-S6K1
immunomodulatory ability are safe and effective in signalling pathway, which provides a basis for the
T2DM patients, and microencapsulated HUC-MSCs clinical application of HUC-MSCs in treating
reduce effector Th1 cells and repair the Treg/Th17 pancreatitis (89). In 2019, HucMSC-ex delivered
ratio (77), suggesting that HUC-MSCs may treat exogenous miR-145-5p to inhibit pancreatic ductal
T1DM by modulating immunity. In addition, HUC- adenocarcinoma progression, suggesting a thera-
MSCs have shown efficacy in other autoimmune peutic role of HUC-MSCs in pancreatic exocrine
diseases, such as T1DM combined with Sjogren diseases (90). Additionally, transplantation of
syndrome (SS) (78, 79). Overall, MSCs may represent HUC-MSCs can effectively alleviate weight loss
a new strategy to treat immune-mediated diseases. symptoms, reduce blood glucose levels, and improve
offspring survival in gestational diabetes mellitus
HUC-MSCs improve insulin resistance (GDM) patients (91). However, it has been shown that
Insulin resistance (IR) is one of the most common GDM adversely affects the proliferative capacity and
and important pathological features of T2DM. MSCs viability of HUC-MSCs (92). Therefore, to address this
can exert immunomodulatory and anti-inflammatory situation, it is crucial to identify conditions that
effects through paracrine effects, thereby increasing improve the survival of HUC-MSCs, reduce
insulin sensitivity and improving insulin resistance in apoptosis, and promote proliferation.
T2DM rats (80). Umbilical cord mesenchymal stem In conclusion, MSC therapy presents a novel
cell-conditioned medium (UC-MSC-CM) may approach for treating DM, displaying substantial
improve IR in C2C12 cells by improving glucose efficacy in both basic and clinical trials. HUC-MSCs
transporter 4 (GLUT4) translocation, insulin exhibit the capacity to migrate towards damaged
signalling pathways, and mitochondrial content and pancreatic islets, facilitated by homing and paracrine
function (81). HUC-MSCs also improve IR by effects, thus assuming a reparative role. Additionally,
modulating the balance between PTEN-mediated they induce differentiation into IPCs, replacing
PI3K/Akt and ERK/MAPK signalling pathways (82). impaired islet β-cells and enabling the secretion of C
UC-MSCs infusion and fasting-mimicking diet (FMD) peptide and insulin. Furthermore, they counteract
synergistically modulate the systemic inflammatory β-cell de-differentiation, thereby safeguarding
microenvironment and improve hyperglycaemia and pancreatic β-cells, and facilitate regeneration of islet
lipid metabolism disorders in T2DM mice (83). β-cells along with structural revitalization, conseq-
Glucagon-like peptide-1 (GLP-1) gene modification of uently enhancing islet β-cell functionality. Their
HUC-MSCs improves fasting glucose, IR, and β-cell impact extends to immune cells, encompassing
function in T2DM mice (84). HUC-MSCs combined macrophages, DCs, NK cells, neutrophils, and T cells,
with liraglutide can downregulate the TLR4/NF-kB thereby exerting immunomodulatory and anti-
inflammatory pathway and oxidative stress while inflammatory properties. This therapeutic modality
improving glucose metabolism and inhibiting islet also ameliorates insulin resistance by targeting
β-cell apoptosis in an ASK1/JNK/BAX pathway- insulin-responsive organs. In summary, the collective
dependent manner in T2DM rats (85, 86). In mechanisms through which HUC-MSCs operate
https://www.medsci.org
Int. J. Med. Sci. 2023, Vol. 20 1497
Table 1. The possible modes of action of MSCs in the treatment of diabetes are discussed in the table.
Mode of action of HUC-MSCs
Mode of action Mechanism References
Homing effects Systemic homing Initial tethering by selectins (22, 23)
Activation by cytokines
Blockade by integrins
Exudation or migration using matrix remodelling agents
Extravasation toward chemokine gradients
Non-systematic homing Directed to the injury site via a chemokine gradient.
Paracrine effects Secrete soluble molecules (KGF, HGF, VEGF, FGF, PGF, MCP-1, IGF-1, Promoting tissue regeneration and angiogenesis (12, 28-30)
EGF, PGE2, IDO, IL-10, IL-6, TGF-β1, NO, HLA-G5, TSG-6, and Promoting ulcer tissue healing and wound healing
neurotrophic factors) Modulating immunity, anti-inflammation, anti-apoptosis,
and cytoprotection.
Release of EVs, such as exosomes and microvesicles Activating the regenerative capacity of islets (31-33)
Improving insulin sensitivity
Reversing peripheral insulin resistance
Attenuating β-cell destruction
Differentiation into Induced to differentiate into IPCs Replace some damaged islet β-cells to secrete C peptide and (40, 45)
IPCs INS
Improve islet β-cell Protection of pancreatic islet beta cells Secreting IL-1Ra to reduce islet injury and reverse β-cell (60, 62)
function dedifferentiation
Promoting the regeneration of pancreatic islet beta cells Induced to differentiate into pβLCs functional pancreatic β (63)
cells
Immunomodulatory Macrophage Suppress inflammation and improve insulin sensitivity by (25, 69-71)
effects secreting IL-6 and IL-10 to promote M2 macrophage
polarization.
DCs, NK cells, neutrophil, and T cells Regulate antigen presentation by DCs (74)
Regulate cytotoxicity of NK
Regulate neutrophil activation
Induce peripheral tolerance in T cells
Improve insulin Liver, fat and skeletal muscle Improve IR in C2C12 cells and improve lipid metabolism (81, 83)
resistance disorders in T2DM mice
HUC-MSCs, Human umbilical cord mesenchymal stem cells; KGF, Keratinocyte growth factor; HGF, Hepatocyte growth factor; VEGF, Vascular endothelial growth factor;
FGF, Fibroblast growth factor; PGF, Placental growth factor; MCP-1, Monocyte chemoattractant protein 1; IGF-1, Insulin-like growth factor 1; EGF, Epidermal growth factor;
PGE2, Prostaglandin E2; IDO, Indoleamine2,3-deoxygenase; IL-10, Interleukin-10; IL-6, Interleukin-6; TGF-β1, Transforming growth factor-β1; NO, Nitric oxide; HLA-G5,
https://www.medsci.org
Int. J. Med. Sci. 2023, Vol. 20 1498
Human leukocyte antigen-G5; TSG-6, Tumor necrosis factor α stimulated gene 6; EVs, Extracellular vehicles; β-cell, Beta cell; IPCs, Insulin-producing cells; INS, Insulin;
IL-1Ra, Interleukin-1 Receptor antagonist; PβLCs, Pancreatic β-like cells; DCs, Dendritic cells; NK cells, Natural killer cell; IR, Insulin resistance; T2DM, Type 2 diabetes
mellitus.
https://www.medsci.org
Int. J. Med. Sci. 2023, Vol. 20 1499
endothelial nitric oxide synthase (eNOS), IGF1, and pro-inflammatory cytokines and proteases, and
basic fibroblast growth factor (bFGF) (125). In reduced levels of various growth factors (139-141).
conclusion, transplantation of HUC-MSCs may be a HUC-MSCs can self-renew, multi-directionally
new potential therapeutic option for DAN. differentiate, and secrete multiple cytokines and
growth factors, and their mechanisms to improve
Diabetic foot disease diabetic wound healing mainly include 1) promoting
Diabetic foot ulcers (DFU) are full-length lesions diabetic wound healing by differentiating into
that occur in the skin of the foot of patients with DM, keratin-forming cells (142); 2) secreting molecules
accompanied by infection and tissue destruction related to wound healing in a paracrine manner
caused by neuropathy and/or peripheral artery (VEGF, PDGF, KGF, TGF-β1, SMA, scavenger
disease (PAD) (126). DFU has a high disability and receptor class B type1 (SR-B1), and platelet endo-
mortality rate, which severely affects the quality of life thelial cell adhesion molecule-1 (PECAM-1/CD31)) to
of patients, shortens life expectancy, and imposes a promote angiogenesis (143-146); 3) regulating the
heavy socioeconomic burden (127, 128). In recent activity, function, and proliferative capacity of
years, HUC-MSCs have achieved good therapeutic vascular endothelial cells by reducing oxidative stress
effects in the treatment of DFU. Zhao et al. found that and inflammatory response, thereby promoting
HUC-MSCs specifically homed to ulcerated tissue and angiogenesis (147, 148); 4) inducing functional
promoted epithelialisation of ulcerated tissue, recovery of vascular endothelial cells by modulating
possibly by stimulating the release of cytokeratin 19 macrophage phenotype (149); and 5) stimulating
from keratin-forming cells and promoting diabetic fibroblast activity and promoting cell
extracellular matrix formation (129). Shi et al. found proliferation, collagen synthesis, and glycosamino-
that HUC-MSCs promoted wound healing in DFU glycan levels, thereby playing a role in skin wound
rats by transdifferentiating, regulating inflammation, healing play a role (150). Moreover, HUC-MSCs may
and providing growth factors that promote angio- be more effective than fibroblasts in stimulating
genesis, cell proliferation, and collagen deposition diabetic wound healing (151, 152). Additionally,
(130). Xia et al. found that HUC-MSCs prevented or HUC-MSCs accelerate wound healing in diabetic rats
cured foot ulcers in DFU rats by reversing the by increasing epidermal and dermal thickness and
neuronal structure and function by upregulating NGF density, accelerating epithelial and collagen
and promoting significant angiogenesis in the femoral regeneration, and increasing angiogenesis (153). Han
nerve-innervated gastrocnemius muscle (131). et al. found that the Wnt signalling pathway
HUC-MSCs induce angiogenesis (132, 133), promote activation promoted the proliferation and
tissue repair and regeneration (134), and reduce differentiation of HUC-MSCs, thereby facilitating the
muscle damage and apoptosis in the ischaemic hind healing of diabetic skin wounds (154). Yue et al. found
limbs of DM mice (135). Transplantation of that c-Jun overexpression promotes the proliferation
HUC-MSCs significantly improved skin temperature, and migration of HUC-MSCs in vitro and accelerates
ankle-arm pressure index, transcutaneous partial diabetic wound closure, re-epithelialization, and
pressure of oxygen, and claudication distance in angiogenesis in vivo (155). The development of new
patients with postoperative diabetic foot disease. This technologies has extensively improved the
is accompanied by a significant increase in therapeutic efficacy of HUC-MSCs. HUC-MSCs can
neovascularization and complete or gradual ulcer improve skin wound healing in diabetic mice by
healing (136). In conclusion, transplantation of combining with Pluronic F127 hydrogel (156),
HUC-MSCs may be a potential strategy for clinical tissue-engineered scaffolds (157, 158), or Cas9-AAV6
application in DFU, although its long-term effects engineering modification (159). Overall, HUC-MSC
remain to be elucidated. transplantation may have a therapeutic effect on
impaired diabetic wound healing; however, its
Impaired wound healing in DM
specific therapeutic modalities and safety need to be
Impaired wound healing is a common DM further explored.
complication. DM is associated with persistent
inflammation and a defective tissue repair response. HUC-MSCs infusion is safe and effective
Impaired angiogenesis is an important factor in for COVID-19 with diabetes
delaying chronic diabetic wound healing. Poorly
healing wounds in DM mice exhibit a persistent Currently, coronavirus disease 2019 (COVID-19)
inflammatory response, a deficiency of M2 is a serious global public health problem and is
macrophages (137, 138), a prolonged accumulation of significantly associated with an increased risk of
pro-inflammatory M1 macrophages, elevated levels of developing DM (160). At the same time, patients with
DM are at a high risk of developing severe COVID-19
https://www.medsci.org
Int. J. Med. Sci. 2023, Vol. 20 1500
infections, have a complex disease process, and have an immunostimulatory state in MSCs, suggesting that
significantly higher mortality rates (161, 162). Severe the metabolic disease environment alters the
COVID-19 is thought to result from the hyper- immunomodulatory efficacy of healthy donor MSCs
inflammatory state and overactive immune response (173). Boland et al. found that culturing HUC-MSCs in
caused by severe acute respiratory syndrome xeno-free conditions attenuated palmitate-induced
coronavirus 2 (SARS-CoV-2) infection, as well as impairment of the immunomodulatory function of
cytokine storm and immune thrombosis. The SARS- HUC-MSCs (174). Additionally, the mode of MSCs
CoV-2 spike glycoprotein binds to angiotensin- administration affects therapeutic efficacy, with
converting enzyme 2 (ACE2), and the serine protease intravenous delivery methods being more effective
transmembrane protease serine 2 (TMPRSS2) initiates than intraperitoneal grafts (175). Local delivery causes
S proteins that can facilitate viral entry into cells, viral MSCs to cluster into "spheroids", thereby altering
replication, and cell-to-cell transmission (163). The gene expression and phenotype. In 2020, researchers
activity of ACE2 is increased in DM mice (164, 165) found that budesonide could act synergistically with
and significantly increased in patients with DM prostaglandin E2 (PGE2) produced by spheroid MSCs
treated with angiotensin-converting enzyme to inhibit T cell proliferation at the PGE2 receptors
inhibitors (ACEI) and angiotensin receptor blockers EP2 and EP4 (176). Moreover, IPCs may be
(ARB) (166), suggesting that patients with DM may be immunogenic and trigger immune responses after
at increased COVID-19 risk. transplantation into the host owing to changes in the
MSCs can achieve immunomodulation by immune microenvironment and immune cell
secreting various cytokines through paracrine infiltration, thus reducing cell survival and further
pathways or by interacting directly with the immune differentiation (177). However, encapsulation of IPCs
cells (167). ACE2 and TMPRSS2 were expressed at with alginate has been shown to avoid graft rejection,
low levels in HUC-MSCs, suggesting that HUC-MSCs which greatly improves the efficacy of allogeneic or
may have the ability to "evade" viral infection and xenogeneic MSCs in the treatment of DM (178).
thus exert immunomodulatory effects (168). Stem cell banking is the most important life
HUC-MSCs reduced the levels of inflammatory resource for human beings, which can provide
molecules associated with the COVID-19 "cytokine high-quality seed cell resources for stem cell therapy.
storm", including interferon-γ (IFNγ), IL1β, IL-6, and By establishing a standardized production process of
TNFα, and regulated upon activation of normal T cell MSCs, it can improve stem cell preparation quality
expressed and secreted factor (RANTES). and promote the sustainable development of stem cell
Additionally, no serious adverse events related to clinical applications. Actively promoting the clinical
HUC-MSC infusion have been observed (169, 170). translation of stem cell therapy and improving the
HUC-MSCs improved respiratory distress and survival rate and efficacy of HUC-MSCs after
reduced inflammatory biomarkers in patients with transplantation will become the top priority of stem
critically ill COVID-19-induced automated resources cell technology research nowadays. With the
directory service (ARDS) (171). Tao et al. found that development of technology, the field of stem cell
UC-MSCs significantly increased pulmonary static research has become a frontier hotspot. The
compliance, maintained a stable partial pressure of implantation of the bioartificial pancreas (179), the
oxygen (PaO2)/fraction of inspired oxygen (FiO2) labelling of nanoparticles (NP) (180), and the
ratio, and improved renal function in critically ill co-microencapsulation of HUC-MSCs/human
COVID-19 patients, suggesting that UC-MSCs pancreatic islet-derived progenitor cells (hIDC) (181)
transplantation may have a positive therapeutic effect may provide new tools for cellular therapy of DM.
in critically ill COVID-19 patients (172). In conclusion, Carboxylic acid-functionalized single-walled carbon
HUC-MSC therapy may be a potential treatment nanotubes (f-SWCNT-COOH) (182) and some
option for DM combined with COVID-19. cytokines (183) can increase the viability and ex vivo
expansion of hematopoietic stem cell (HSC) and/or
Opportunities and challenges hematopoietic stem progenitor cell (HSPC). These
HUC-MSCs have shown impressive results in studies provide new perspectives for developing DM
treating DM and its complications, but most studies cell transplantation therapies based on HUC-MSCs.
are still in the preclinical stage. Improving the We believe that the effectiveness of HUC-MSC
survival and efficacy of HUC-MSCs after transplanta- therapy will be greatly improved by applying
tion in a challenging metabolic environment may be advanced technologies such as gene modification,
an interesting topic in the future, as elevated nanotechnology, magnetic targeting technology, and
palmitate levels in the sera of obese and T2DM tissue engineering technology. We believe that soon,
patients lead to a shift from an immunosuppressive to HUC-MSCs may provide a better solution for the
https://www.medsci.org
Int. J. Med. Sci. 2023, Vol. 20 1501
Figure 2. This figure illustrates the broad effect of HUC-MSCs on DM and its complications, as well as the therapeutic effect of HUC-MSCs on diabetic patients infected with
COVID-19. COVID-19: coronavirus disease 2019.
clinical treatment of DM and its complications, and research is needed to improve the homing rate,
thus can bring new hope to a greater extent in DM survival rate, efficacy, and safety of MSCs after
patients worldwide. transplantation. With the gradual maturation of
technology and theory, the fundamental treatment of
Conclusions diabetes will usher in a greater breakthrough. We
The DM epidemic and its complications pose a believe that HUC-MSCs transplantation can provide
major threat to global health, accompanied by high more options for the management of DM and its
morbidity and mortality. Currently, there are many complications and bring longer-term benefits to
ways to treat DM, such as traditional oral patients.
hypoglycaemic therapy and insulin injections, but
they can only temporarily control blood glucose levels Abbreviations
and cannot cure diabetes, and have insufficient DM: diabetes mellitus
control over diabetic complications, in addition to IDF: international diabetes federation
long-term use of hypoglycaemic drugs or insulin T1DM: type 1 diabetes mellitus
injections, which significantly reduces patient T2DM: type 2 diabetes mellitus
compliance. Recently, regenerative medicine with MSCs: mesenchymal stem cells
MSCs treatment as the core has provided new ideas HUC-MSCs: human umbilical cord
and possible development directions for DM mesenchymal stem cells
treatment. The characteristics of HUC-MSCs, such as UCB: umbilical cord blood
abundant source, less ethical controversy, lower risk WJ: wharton's jelly
of infection, higher proliferation and differentiation PDGF: platelet derived growth factor
ability, and very low immunogenicity, make them BM-MSCs: bone marrow-derived mesenchymal
stand out among MSCs of different tissue sources. We stem cells
mainly describe the application of HUC-MSCs in DM PU-MSCs: pulp-derived mesenchymal stem cells
and its complications. HUC-MSCs transplantation is AD-MSCs: adipose tissue-derived mesenchymal
expected to be an efficient and ideal treatment for DM stem cells
and its complications, and its application area will β-cell: beta cell
gradually expand (Fig. 2). Currently, HUC-MSCs DiI: 1,1'-dioctadecyl-3,3,3',3'-tetramethylindo-
therapy is still in the exploration stage, and further carbocyanine perchlorate
https://www.medsci.org
Int. J. Med. Sci. 2023, Vol. 20 1502
https://www.medsci.org
Int. J. Med. Sci. 2023, Vol. 20 1503
NP: nanoparticles 12. Xie Q, Liu R, Jiang J, Peng J, Yang C, Zhang W. et al. What is the impact of
human umbilical cord mesenchymal stem cell transplantation on clinical
hIDC: human pancreatic islet-derived progenitor treatment? Stem cell research & therapy. 2020;11(1):519.
13. Simões IN, Boura JS, dos Santos F, Andrade PZ, Cardoso CM, Gimble JM. et al.
cells Human mesenchymal stem cells from the umbilical cord matrix: successful
f-SWCNT-COOH: carboxylic acid functionalized isolation and ex vivo expansion using serum-/xeno-free culture media.
Biotechnology journal. 2013;8(4):448-58.
single walled carbon nanotubes 14. Choudhery MS, Badowski M, Muise A, Harris DT. Utility of cryopreserved
HSC: hematopoietic stem cell umbilical cord tissue for regenerative medicine. Current stem cell research &
therapy. 2013;8(5):370-80.
HSPC: hematopoietic stem progenitor cell 15. Li J, Xu SQ, Zhao YM, Yu S, Ge LH, Xu BH. Comparison of the biological
characteristics of human mesenchymal stem cells derived from exfoliated
Acknowledgements deciduous teeth, bone marrow, gingival tissue, and umbilical cord. Molecular
medicine reports. 2018;18(6):4969-77.
Figures for this work were created in 16. Deans RJ, Moseley AB. Mesenchymal stem cells: biology and potential clinical
uses. Experimental hematology. 2000;28(8):875-84.
BioRender.com and are gratefully acknowledged. 17. Rao MS, Mattson MP. Stem cells and aging: expanding the possibilities.
Mechanisms of ageing and development. 2001;122(7):713-34.
Funding 18. Wu LF, Wang NN, Liu YS, Wei X. Differentiation of Wharton's jelly primitive
stromal cells into insulin-producing cells in comparison with bone marrow
The financial support for this work from the mesenchymal stem cells. Tissue engineering Part A. 2009;15(10):2865-73.
19. Ma Y, Wang L, Yang S, Liu D, Zeng Y, Lin L. et al. The tissue origin of human
National Natural Science Foundation of China (Grant mesenchymal stem cells dictates their therapeutic efficacy on glucose and lipid
No. 81471028) and Jilin Provincial Department of metabolic disorders in type II diabetic mice. Stem cell research & therapy.
2021;12(1):385.
science and technology (Grant No. 3D5223990429) is 20. Wang H, Qiu X, Ni P, Qiu X, Lin X, Wu W. et al. Immunological characteristics
of human umbilical cord mesenchymal stem cells and the therapeutic effects
gratefully acknowledged. of their transplantion on hyperglycemia in diabetic rats. International journal
of molecular medicine. 2014;33(2):263-70.
Author contributions 21. Zang L, Hao H, Liu J, Li Y, Han W, Mu Y. Mesenchymal stem cell therapy in
type 2 diabetes mellitus. Diabetology & metabolic syndrome. 2017;9:36.
Luyao Li contributed to the conceptualization 22. Liesveld JL, Sharma N, Aljitawi OS. Stem cell homing: From physiology to
and wrote the manuscript.Jicui Li contributed to therapeutics. Stem cells (Dayton, Ohio). 2020;38(10):1241-53.
23. Ullah M, Liu DD, Thakor AS. Mesenchymal Stromal Cell Homing:
reviewing the draft and making necessary Mechanisms and Strategies for Improvement. iScience. 2019;15:421-38.
modifcations, and Haifei Guan participated in 24. Maldonado M, Huang T, Yang L, Xu L, Ma L. Human umbilical cord Wharton
jelly cells promote extra-pancreatic insulin formation and repair of renal
drawing the figures. Chuan Zhang,Hisashi Oishi and damage in STZ-induced diabetic mice. Cell communication and signaling :
CCS. 2017;15(1):43.
Satoru Takahashi designed the study. All authors 25. Yin Y, Hao H, Cheng Y, Gao J, Liu J, Xie Z. et al. The homing of human
read and approved the final manuscript. umbilical cord-derived mesenchymal stem cells and the subsequent
modulation of macrophage polarization in type 2 diabetic mice. International
immunopharmacology. 2018;60:235-45.
Competing Interests 26. Keshtkar S, Azarpira N, Ghahremani MH. Mesenchymal stem cell-derived
extracellular vesicles: novel frontiers in regenerative medicine. Stem cell
The authors have declared that no competing research & therapy. 2018;9(1):63.
interest exists. 27. Kusuma GD, Carthew J, Lim R, Frith JE. Effect of the Microenvironment on
Mesenchymal Stem Cell Paracrine Signaling: Opportunities to Engineer the
Therapeutic Effect. Stem cells and development. 2017;26(9):617-31.
References 28. Vija L, Farge D, Gautier JF, Vexiau P, Dumitrache C, Bourgarit A. et al.
Mesenchymal stem cells: Stem cell therapy perspectives for type 1 diabetes.
1. Saeedi P, Petersohn I, Salpea P, Malanda B, Karuranga S, Unwin N. et al. Diabetes & metabolism. 2009;35(2):85-93.
Global and regional diabetes prevalence estimates for 2019 and projections for 29. Stiner R, Alexander M, Liu G, Liao W, Liu Y, Yu J. et al. Transplantation of
2030 and 2045: Results from the International Diabetes Federation Diabetes stem cells from umbilical cord blood as therapy for type I diabetes. Cell and
Atlas, 9(th) edition. Diabetes research and clinical practice. 2019;157:107843. tissue research. 2019;378(2):155-62.
2. Ridler C. Diabetes: Islet transplantation for T1DM. Nature reviews 30. Khubutiya MS, Vagabov AV, Temnov AA, Sklifas AN. Paracrine mechanisms
Endocrinology. 2016;12(7):373. of proliferative, anti-apoptotic and anti-inflammatory effects of mesenchymal
3. Wu Y, Ding Y, Tanaka Y, Zhang W. Risk factors contributing to type 2 diabetes stromal cells in models of acute organ injury. Cytotherapy. 2014;16(5):579-85.
and recent advances in the treatment and prevention. International journal of 31. Sharma R, Kumari M, Mishra S, Chaudhary DK, Kumar A, Avni B. et al.
medical sciences. 2014;11(11):1185-200. Exosomes Secreted by Umbilical Cord Blood-Derived Mesenchymal Stem Cell
4. Papatheodorou K, Papanas N, Banach M, Papazoglou D, Edmonds M. Attenuate Diabetes in Mice. Journal of diabetes research. 2021;2021:9534574.
Complications of Diabetes 2016. Journal of diabetes research. 32. Chen MT, Zhao YT, Zhou LY, Li M, Zhang Q, Han Q. et al. Exosomes Derived
2016;2016:6989453. from Human Umbilical Cord Mesenchymal Stem Cells Enhance Insulin
5. Ali MK, Pearson-Stuttard J, Selvin E, Gregg EW. Interpreting global trends in Sensitivity in Insulin Resistant Human Adipocytes. Current medical science.
type 2 diabetes complications and mortality. Diabetologia. 2022;65(1):3-13. 2021;41(1):87-93.
6. DelaRosa O, Lombardo E. Modulation of adult mesenchymal stem cells 33. Sun Y, Shi H, Yin S, Ji C, Zhang X, Zhang B. et al. Human Mesenchymal Stem
activity by toll-like receptors: implications on therapeutic potential. Mediators Cell Derived Exosomes Alleviate Type 2 Diabetes Mellitus by Reversing
of inflammation. 2010;2010:865601. Peripheral Insulin Resistance and Relieving β-Cell Destruction. ACS nano.
7. Davies JE, Walker JT, Keating A. Concise Review: Wharton's Jelly: The Rich, 2018;12(8):7613-28.
but Enigmatic, Source of Mesenchymal Stromal Cells. Stem cells translational 34. Yap SK, Tan KL, Abd Rahaman NY, Saulol Hamid NF, Ooi J, Tor YS. et al.
medicine. 2017;6(7):1620-30. Human Umbilical Cord Mesenchymal Stem Cell-Derived Small Extracellular
8. Romanov YA, Svintsitskaya VA, Smirnov VN. Searching for alternative Vesicles Ameliorated Insulin Resistance in Type 2 Diabetes Mellitus Rats.
sources of postnatal human mesenchymal stem cells: candidate MSC-like cells Pharmaceutics. 2022;14(3).
from umbilical cord. Stem cells (Dayton, Ohio). 2003;21(1):105-10. 35. Chen J, Chen J, Cheng Y, Fu Y, Zhao H, Tang M. et al. Mesenchymal stem
9. Kita K, Gauglitz GG, Phan TT, Herndon DN, Jeschke MG. Isolation and cell-derived exosomes protect beta cells against hypoxia-induced apoptosis
characterization of mesenchymal stem cells from the sub-amniotic human via miR-21 by alleviating ER stress and inhibiting p38 MAPK
umbilical cord lining membrane. Stem cells and development. phosphorylation. Stem cell research & therapy. 2020;11(1):97.
2010;19(4):491-502. 36. Mohammadi MR, Rodriguez SM, Luong JC, Li S, Cao R, Alshetaiwi H. et al.
10. Grzywocz Z, Pius-Sadowska E, Klos P, Gryzik M, Wasilewska D, Exosome loaded immunomodulatory biomaterials alleviate local immune
Aleksandrowicz B. et al. Growth factors and their receptors derived from response in immunocompetent diabetic mice post islet xenotransplantation.
human amniotic cells in vitro. Folia histochemica et cytobiologica. Communications biology. 2021;4(1):685.
2014;52(3):163-70. 37. Moshrefi M, Yari N, Nabipour F, Bazrafshani MR, Nematollahi-mahani SN.
11. Harris DT. Umbilical cord tissue mesenchymal stem cells: characterization and Transplantation of differentiated umbilical cord mesenchymal cells under
clinical applications. Current stem cell research & therapy. 2013;8(5):394-9. kidney capsule for control of type I diabetes in rat. Tissue & cell.
2015;47(4):395-405.
https://www.medsci.org
Int. J. Med. Sci. 2023, Vol. 20 1504
38. Wang HS, Shyu JF, Shen WS, Hsu HC, Chi TC, Chen CP. et al. Transplantation 62. Li B, Cheng Y, Yin Y, Xue J, Yu S, Gao J. et al. Reversion of early- and late-stage
of insulin-producing cells derived from umbilical cord stromal mesenchymal β-cell dedifferentiation by human umbilical cord-derived mesenchymal stem
stem cells to treat NOD mice. Cell transplantation. 2011;20(3):455-66. cells in type 2 diabetic mice. Cytotherapy. 2021;23(6):510-20.
39. Montanucci P, Pescara T, Alunno A, Bistoni O, Basta G, Calafiore R. Remission 63. Zhang T, Wang H, Wang T, Wei C, Jiang H, Jiang S. et al. Pax4 synergistically
of hyperglycemia in spontaneously diabetic NOD mice upon transplant of acts with Pdx1, Ngn3 and MafA to induce HuMSCs to differentiate into
microencapsulated human umbilical cord Wharton jelly-derived functional pancreatic β-cells. Experimental and therapeutic medicine.
mesenchymal stem cells (hUCMS). Xenotransplantation. 2019;26(2):e12476. 2019;18(4):2592-8.
40. Prabakar KR, Domínguez-Bendala J, Molano RD, Pileggi A, Villate S, Ricordi 64. Wei L, Zhang L, Yang L, Wang X, Zhao C, Zhao D. Protective Effect of
C. et al. Generation of glucose-responsive, insulin-producing cells from Mesenchymal Stem Cells on Isolated Islets Survival and Against Hypoxia
human umbilical cord blood-derived mesenchymal stem cells. Cell Associated With the HIF-1α/PFKFB3 Pathway. Cell transplantation.
transplantation. 2012;21(6):1321-39. 2022;31:9636897211073127.
41. Su X, Fang S, Zhang D, Zhang Q, He Y, Lu X. et al. Quantitative Raman 65. Zhou Y, Hu Q, Chen F, Zhang J, Guo J, Wang H. et al. Human umbilical cord
spectral changes of the differentiation of mesenchymal stem cells into islet-like matrix-derived stem cells exert trophic effects on β-cell survival in diabetic rats
cells by biochemical component analysis and multiple peak fitting. Journal of and isolated islets. Disease models & mechanisms. 2015;8(12):1625-33.
biomedical optics. 2015;20(12):125002. 66. Bao Y, Zhao Z, Gao H. Effect of hTIMP-1 overexpression in human umbilical
42. Phuc PV, Nhung TH, Loan DT, Chung DC, Ngoc PK. Differentiating of cord mesenchymal stem cells on the repair of pancreatic islets in type-1
banked human umbilical cord blood-derived mesenchymal stem cells into diabetic mice. Cell biology international. 2021;45(5):1038-49.
insulin-secreting cells. In vitro cellular & developmental biology Animal. 67. Lu J, Shen SM, Ling Q, Wang B, Li LR, Zhang W. et al. One repeated
2011;47(1):54-63. transplantation of allogeneic umbilical cord mesenchymal stromal cells in type
43. Wang HW, Lin LM, He HY, You F, Li WZ, Huang TH. et al. Human umbilical 1 diabetes: an open parallel controlled clinical study. Stem cell research &
cord mesenchymal stem cells derived from Wharton's jelly differentiate into therapy. 2021;12(1):340.
insulin-producing cells in vitro. Chinese medical journal. 2011;124(10):1534-9. 68. Hu J, Wang F, Sun R, Wang Z, Yu X, Wang L. et al. Effect of combined therapy
44. Hu YH, Wu DQ, Gao F, Li GD, Yao L, Zhang XC. A secretory function of of human Wharton's jelly-derived mesenchymal stem cells from umbilical
human insulin-producing cells in vivo. Hepatobiliary & pancreatic diseases cord with sitagliptin in type 2 diabetic rats. Endocrine. 2014;45(2):279-87.
international : HBPD INT. 2009;8(3):255-60. 69. Xie Z, Hao H, Tong C, Cheng Y, Liu J, Pang Y. et al. Human umbilical
45. Yu YB, Bian JM, Gu DH. Transplantation of insulin-producing cells to treat cord-derived mesenchymal stem cells elicit macrophages into an
diabetic rats after 90% pancreatectomy. World journal of gastroenterology. anti-inflammatory phenotype to alleviate insulin resistance in type 2 diabetic
2015;21(21):6582-90. rats. Stem cells (Dayton, Ohio). 2016;34(3):627-39.
46. Chao KC, Chao KF, Chen CF, Liu SH. A novel human stem cell coculture 70. Xue J, Gao J, Gu Y, Wang A, Yu S, Li B. et al. Human umbilical cord-derived
system that maintains the survival and function of culture islet-like cell mesenchymal stem cells alleviate insulin resistance in diet-induced obese mice
clusters. Cell transplantation. 2008;17(6):657-64. via an interaction with splenocytes. Stem cell research & therapy.
47. Wang G, Li Y, Wang Y, Dong Y, Wang FS, Ding Y. et al. Roles of the co-culture 2022;13(1):109.
of human umbilical cord Wharton's jelly-derived mesenchymal stem cells with 71. Yin Y, Hao H, Cheng Y, Zang L, Liu J, Gao J. et al. Human umbilical
rat pancreatic cells in the treatment of rats with diabetes mellitus. cord-derived mesenchymal stem cells direct macrophage polarization to
Experimental and therapeutic medicine. 2014;8(5):1389-96. alleviate pancreatic islets dysfunction in type 2 diabetic mice. Cell death &
48. Seyedi F, Farsinejad A, Moshrefi M, Nematollahi-Mahani SN. In vitro disease. 2018;9(7):760.
evaluation of different protocols for the induction of mesenchymal stem cells 72. Xue J, Cheng Y, Hao H, Gao J, Yin Y, Yu S. et al. Low-Dose Decitabine Assists
to insulin-producing cells. In vitro cellular & developmental biology Animal. Human Umbilical Cord-Derived Mesenchymal Stem Cells in Protecting β
2015;51(8):866-78. Cells via the Modulation of the Macrophage Phenotype in Type 2 Diabetic
49. Gao F, Wu DQ, Hu YH, Jin GX. Extracellular matrix gel is necessary for in Mice. Stem cells international. 2020;2020:4689798.
vitro cultivation of insulin producing cells from human umbilical cord blood 73. Jiang W, Xu J. Immune modulation by mesenchymal stem cells. Cell
derived mesenchymal stem cells. Chinese medical journal. 2008;121(9):811-8. proliferation. 2020;53(1):e12712.
50. He D, Wang J, Gao Y, Zhang Y. Differentiation of PDX1 gene-modified human 74. Uccelli A, Moretta L, Pistoia V. Mesenchymal stem cells in health and disease.
umbilical cord mesenchymal stem cells into insulin-producing cells in vitro. Nature reviews Immunology. 2008;8(9):726-36.
International journal of molecular medicine. 2011;28(6):1019-24. 75. Li XY, Zheng ZH, Li XY, Guo J, Zhang Y, Li H. et al. Treatment of foot disease
51. Wang XL, Hu P, Guo XR, Yan D, Yuan Y, Yan SR. et al. Reprogramming in patients with type 2 diabetes mellitus using human umbilical cord blood
human umbilical cord mesenchymal stromal cells to islet-like cells with the mesenchymal stem cells: response and correction of immunological
use of in vitro-synthesized pancreatic-duodenal homebox 1 messenger RNA. anomalies. Current pharmaceutical design. 2013;19(27):4893-9.
Cytotherapy. 2014;16(11):1519-27. 76. Kong D, Zhuang X, Wang D, Qu H, Jiang Y, Li X. et al. Umbilical cord
52. Hu YH, Wu DQ, Gao F, Li GD, Zhang XC. Notch signaling: a novel regulating mesenchymal stem cell transfusion ameliorated hyperglycemia in patients
differentiation mechanism of human umbilical cord blood-derived with type 2 diabetes mellitus. Clinical laboratory. 2014;60(12):1969-76.
mesenchymal stem cells into insulin-producing cells in vitro. Chinese medical 77. Montanucci P, Alunno A, Basta G, Bistoni O, Pescara T, Caterbi S. et al.
journal. 2010;123(5):606-14. Restoration of t cell substes of patients with type 1 diabetes mellitus by
53. Qu H, Liu X, Ni Y, Jiang Y, Feng X, Xiao J. et al. Laminin 411 acts as a potent microencapsulated human umbilical cord Wharton jelly-derived
inducer of umbilical cord mesenchymal stem cell differentiation into mesenchymal stem cells: An in vitro study. Clinical immunology (Orlando,
insulin-producing cells. Journal of translational medicine. 2014;12:135. Fla). 2016;163:34-41.
54. Sun B, Meng XH, Liu R, Yan S, Xiao ZD. Mechanism study for hypoxia 78. Qi J, Tang X, Li W, Chen W, Yao G, Sun L. Mesenchymal stem cells inhibited
induced differentiation of insulin-producing cells from umbilical cord the differentiation of MDSCs via COX2/PGE2 in experimental sialadenitis.
blood-derived mesenchymal stem cells. Biochemical and biophysical research Stem cell research & therapy. 2020;11(1):325.
communications. 2015;466(3):444-9. 79. Liu Y, Li C, Wang S, Guo J, Guo J, Fu J. et al. Human umbilical cord
55. Chandravanshi B, Bhonde R. Small molecules exert anti-apoptotic effect and mesenchymal stem cells confer potent immunosuppressive effects in Sjögren's
reduce oxidative stress augmenting insulin secretion in stem cells engineered syndrome by inducing regulatory T cells. Modern rheumatology.
islets against hypoxia. European journal of pharmacology. 2016;791:424-32. 2021;31(1):186-96.
56. Tsai PJ, Wang HS, Shyr YM, Weng ZC, Tai LC, Shyu JF. et al. Transplantation 80. Sun X, Hao H, Han Q, Song X, Liu J, Dong L. et al. Human umbilical
of insulin-producing cells from umbilical cord mesenchymal stem cells for the cord-derived mesenchymal stem cells ameliorate insulin resistance by
treatment of streptozotocin-induced diabetic rats. Journal of biomedical suppressing NLRP3 inflammasome-mediated inflammation in type 2 diabetes
science. 2012;19(1):47. rats. Stem cell research & therapy. 2017;8(1):241.
57. Seyedi F, Farsinejad A, Nematollahi-Mahani SA, Eslaminejad T, 81. Kim KS, Choi YK, Kim MJ, Hwang JW, Min K, Jung SY. et al. Umbilical
Nematollahi-Mahani SN. Suspension Culture Alters Insulin Secretion in Cord-Mesenchymal Stem Cell-Conditioned Medium Improves Insulin
Induced Human Umbilical Cord Matrix-Derived Mesenchymal Cells. Cell Resistance in C2C12 Cell. Diabetes & metabolism journal. 2021;45(2):260-9.
journal. 2016;18(1):52-61. 82. Chen G, Fan XY, Zheng XP, Jin YL, Liu Y, Liu SC. Human umbilical
58. Belame Shivakumar S, Bharti D, Baregundi Subbarao R, Park JM, Son YB, cord-derived mesenchymal stem cells ameliorate insulin resistance via
Ullah I. et al. Pancreatic endocrine-like cells differentiated from human PTEN-mediated crosstalk between the PI3K/Akt and Erk/MAPKs signaling
umbilical cords Wharton's jelly mesenchymal stem cells using small pathways in the skeletal muscles of db/db mice. Stem cell research & therapy.
molecules. Journal of cellular physiology. 2019;234(4):3933-47. 2020;11(1):401.
59. Cinti F, Bouchi R, Kim-Muller JY, Ohmura Y, Sandoval PR, Masini M. et al. 83. Zhao N, Gao YF, Bao L, Lei J, An HX, Pu FX. et al. Glycemic control by
Evidence of β-Cell Dedifferentiation in Human Type 2 Diabetes. The Journal umbilical cord-derived mesenchymal stem cells promotes effects of
of clinical endocrinology and metabolism. 2016;101(3):1044-54. fasting-mimicking diet on type 2 diabetic mice. Stem cell research & therapy.
60. Wang L, Liu T, Liang R, Wang G, Liu Y, Zou J. et al. Mesenchymal stem cells 2021;12(1):395.
ameliorate β cell dysfunction of human type 2 diabetic islets by reversing β cell 84. Chang Y, Dong M, Wang Y, Yu H, Sun C, Jiang X. et al. GLP-1 Gene-Modified
dedifferentiation. EBioMedicine. 2020;51:102615. Human Umbilical Cord Mesenchymal Stem Cell Line Improves Blood Glucose
61. Ortiz LA, Dutreil M, Fattman C, Pandey AC, Torres G, Go K. et al. Interleukin Level in Type 2 Diabetic Mice. Stem cells international. 2019;2019:4961865.
1 receptor antagonist mediates the antiinflammatory and antifibrotic effect of 85. Xu X, Wang W, Lin L, Chen P. Liraglutide in combination with human
mesenchymal stem cells during lung injury. Proceedings of the National umbilical cord mesenchymal stem cell could improve liver lesions by
Academy of Sciences of the United States of America. 2007;104(26):11002-7.
https://www.medsci.org
Int. J. Med. Sci. 2023, Vol. 20 1505
modulating TLR4/NF-kB inflammatory pathway and oxidative stress in effects via transmitting microRNA-18b in rats with diabetic retinopathy.
T2DM/NAFLD rats. Tissue & cell. 2020;66:101382. International immunopharmacology. 2021;101(Pt B):108234.
86. Wang W, Wu RD, Chen P, Xu XJ, Shi XZ, Huang LH. et al. Liraglutide 110. Zhang W, Wang Y, Kong Y. Exosomes Derived From Mesenchymal Stem Cells
combined with human umbilical cord mesenchymal stem cell transplantation Modulate miR-126 to Ameliorate Hyperglycemia-Induced Retinal
inhibits beta-cell apoptosis via mediating the ASK1/JNK/BAX pathway in Inflammation Via Targeting HMGB1. Investigative ophthalmology & visual
rats with type 2 diabetes. Diabetes/metabolism research and reviews. science. 2019;60(1):294-303.
2020;36(2):e3212. 111. Yong M, Kaste M. Dynamic of hyperglycemia as a predictor of stroke outcome
87. Hu J, Yu X, Wang Z, Wang F, Wang L, Gao H. et al. Long term effects of the in the ECASS-II trial. Stroke. 2008;39(10):2749-55.
implantation of Wharton's jelly-derived mesenchymal stem cells from the 112. Hankey GJ, Jamrozik K, Broadhurst RJ, Forbes S, Burvill PW, Anderson CS. et
umbilical cord for newly-onset type 1 diabetes mellitus. Endocrine journal. al. Long-term risk of first recurrent stroke in the Perth Community Stroke
2013;60(3):347-57. Study. Stroke. 1998;29(12):2491-500.
88. Yang B, Bai B, Liu CX, Wang SQ, Jiang X, Zhu CL. et al. Effect of umbilical cord 113. Tuomilehto J, Rastenyte D, Jousilahti P, Sarti C, Vartiainen E. Diabetes mellitus
mesenchymal stem cells on treatment of severe acute pancreatitis in rats. as a risk factor for death from stroke. Prospective study of the middle-aged
Cytotherapy. 2013;15(2):154-62. Finnish population. Stroke. 1996;27(2):210-5.
89. Kong L, Xu X, Zhang H, Zhou Y, Huang H, Chen B. et al. Human umbilical 114. Chen J, Cui X, Zacharek A, Cui Y, Roberts C, Chopp M. White matter damage
cord-derived mesenchymal stem cells improve chronic pancreatitis in rats via and the effect of matrix metalloproteinases in type 2 diabetic mice after stroke.
the AKT-mTOR-S6K1 signaling pathway. Bioengineered. 2021;12(1):1986-96. Stroke. 2011;42(2):445-52.
90. Ding Y, Cao F, Sun H, Wang Y, Liu S, Wu Y. et al. Exosomes derived from 115. Newman MB, Davis CD, Kuzmin-Nichols N, Sanberg PR. Human umbilical
human umbilical cord mesenchymal stromal cells deliver exogenous cord blood (HUCB) cells for central nervous system repair. Neurotoxicity
miR-145-5p to inhibit pancreatic ductal adenocarcinoma progression. Cancer research. 2003;5(5):355-68.
letters. 2019;442:351-61. 116. Chen J, Sanberg PR, Li Y, Wang L, Lu M, Willing AE. et al. Intravenous
91. Wu D, Zou S, Chen H, Li X, Xu Y, Zuo Q. et al. Transplantation routes affect administration of human umbilical cord blood reduces behavioral deficits
the efficacy of human umbilical cord mesenchymal stem cells in a rat GDM after stroke in rats. Stroke. 2001;32(11):2682-8.
model. Clinica chimica acta; international journal of clinical chemistry. 117. Lin CH, Lee HT, Lee SD, Lee W, Cho CW, Lin SZ. et al. Role of
2017;475:137-46. HIF-1α-activated Epac1 on HSC-mediated neuroplasticity in stroke model.
92. Wajid N, Naseem R, Anwar SS, Awan SJ, Ali M, Javed S. et al. The effect of Neurobiology of disease. 2013;58:76-91.
gestational diabetes on proliferation capacity and viability of human umbilical 118. Chen J, Ning R, Zacharek A, Cui C, Cui X, Yan T. et al. MiR-126 Contributes to
cord-derived stromal cells. Cell and tissue banking. 2015;16(3):389-97. Human Umbilical Cord Blood Cell-Induced Neurorestorative Effects After
93. An X, Liao G, Chen Y, Luo A, Liu J, Yuan Y. et al. Intervention for early Stroke in Type-2 Diabetic Mice. Stem cells (Dayton, Ohio). 2016;34(1):102-13.
diabetic nephropathy by mesenchymal stem cells in a preclinical nonhuman 119. Yan T, Venkat P, Ye X, Chopp M, Zacharek A, Ning R. et al. HUCBCs increase
primate model. Stem cell research & therapy. 2019;10(1):363. angiopoietin 1 and induce neurorestorative effects after stroke in T1DM rats.
94. Fang TC, Pang CY, Chiu SC, Ding DC, Tsai RK. Renoprotective effect of CNS neuroscience & therapeutics. 2014;20(10):935-44.
human umbilical cord-derived mesenchymal stem cells in immunodeficient 120. Yan T, Venkat P, Chopp M, Zacharek A, Ning R, Cui Y. et al. Neurorestorative
mice suffering from acute kidney injury. PloS one. 2012;7(9):e46504. Therapy of Stroke in Type 2 Diabetes Mellitus Rats Treated With Human
95. Xiang E, Han B, Zhang Q, Rao W, Wang Z, Chang C. et al. Human umbilical Umbilical Cord Blood Cells. Stroke. 2015;46(9):2599-606.
cord-derived mesenchymal stem cells prevent the progression of early diabetic 121. Vinik AI, Maser RE, Mitchell BD, Freeman R. Diabetic autonomic neuropathy.
nephropathy through inhibiting inflammation and fibrosis. Stem cell research Diabetes care. 2003;26(5):1553-79.
& therapy. 2020;11(1):336. 122. Yuan Z, Tang Z, He C, Tang W. Diabetic cystopathy: A review. Journal of
96. Xian Y, Lin Y, Cao C, Li L, Wang J, Niu J. et al. Protective effect of umbilical diabetes. 2015;7(4):442-7.
cord mesenchymal stem cells combined with resveratrol against renal 123. WenBo W, Fei Z, YiHeng D, Wei W, TingMang Y, WenHao Z. et al. Human
podocyte damage in NOD mice. Diabetes research and clinical practice. Umbilical Cord Mesenchymal Stem Cells Overexpressing Nerve Growth
2019;156:107755. Factor Ameliorate Diabetic Cystopathy in Rats. Neurochemical research.
97. Wang Y, Liu J, Zhang Q, Wang W, Liu Q, Liu S. et al. Human umbilical cord 2017;42(12):3537-47.
mesenchymal stem cells attenuate podocyte injury under high glucose via 124. Shin JH, Ryu CM, Ju H, Yu HY, Song S, Hong KS. et al. Therapeutic Efficacy of
TLR2 and TLR4 signaling. Diabetes research and clinical practice. Human Embryonic Stem Cell-Derived Multipotent Stem/Stromal Cells in
2021;173:108702. Diabetic Detrusor Underactivity: A Preclinical Study. Journal of clinical
98. Nie P, Bai X, Lou Y, Zhu Y, Jiang S, Zhang L. et al. Human umbilical cord medicine. 2020;9(9).
mesenchymal stem cells reduce oxidative damage and apoptosis in diabetic 125. Wu JH, Wang DY, Sheng L, Qian WQ, Xia SJ, Jiang Q. Human umbilical cord
nephropathy by activating Nrf2. Stem cell research & therapy. 2021;12(1):450. Wharton's jelly-derived mesenchymal stem cell transplantation could improve
99. Park JH, Hwang I, Hwang SH, Han H, Ha H. Human umbilical cord diabetic intracavernosal pressure. Asian journal of andrology.
blood-derived mesenchymal stem cells prevent diabetic renal injury through 2022;24(2):171-5.
paracrine action. Diabetes research and clinical practice. 2012;98(3):465-73. 126. Subrata SA, Phuphaibul R. Diabetic foot ulcer care: a concept analysis of the
100. Park JH, Park J, Hwang SH, Han H, Ha H. Delayed treatment with human term integrated into nursing practice. Scandinavian journal of caring sciences.
umbilical cord blood-derived stem cells attenuates diabetic renal injury. 2019;33(2):298-310.
Transplantation proceedings. 2012;44(4):1123-6. 127. American Diabetes Association. Standards of medical care in diabetes--2014.
101. Chang JW, Hung SP, Wu HH, Wu WM, Yang AH, Tsai HL. et al. Therapeutic Diabetes care. 2014;37 Suppl 1:S14-80.
effects of umbilical cord blood-derived mesenchymal stem cell transplantation 128. Santema KTB, Stoekenbroek RM, Koelemay MJW, Reekers JA, van Dortmont
in experimental lupus nephritis. Cell transplantation. 2011;20(2):245-57. LMC, Oomen A. et al. Hyperbaric Oxygen Therapy in the Treatment of
102. Yu C, Yang K, Meng X, Cao B, Wang F. Downregulation of Long Noncoding Ischemic Lower- Extremity Ulcers in Patients With Diabetes: Results of the
RNA MIAT in the Retina of Diabetic Rats with Tail-vein Injection of Human DAMO(2)CLES Multicenter Randomized Clinical Trial. Diabetes care.
Umbilical-cord Mesenchymal Stem Cells. International journal of medical 2018;41(1):112-9.
sciences. 2020;17(5):591-8. 129. Zhao QS, Xia N, Zhao N, Li M, Bi CL, Zhu Q. et al. Localization of human
103. Chen SN, Xu ZG, Ma YX, Chen S, He GH, Han M. et al. Protective effect of mesenchymal stem cells from umbilical cord blood and their role in repair of
LIF-huMSCs on the retina of diabetic model rats. International journal of diabetic foot ulcers in rats. International journal of biological sciences.
ophthalmology. 2021;14(10):1508-17. 2013;10(1):80-9.
104. Gao X, He GH, Zhang XT, Chen S. Protective effect of human umbilical cord 130. Shi R, Lian W, Jin Y, Cao C, Han S, Yang X. et al. Role and effect of
mesenchymal stem cell-derived exosomes on rat retinal neurons in vein-transplanted human umbilical cord mesenchymal stem cells in the repair
hyperglycemia through the brain-derived neurotrophic factor/TrkB pathway. of diabetic foot ulcers in rats. Acta biochimica et biophysica Sinica.
International journal of ophthalmology. 2021;14(11):1683-9. 2020;52(6):620-30.
105. Zhang W, Wang Y, Kong J, Dong M, Duan H, Chen S. Therapeutic efficacy of 131. Xia N, Xu JM, Zhao N, Zhao QS, Li M, Cheng ZF. Human mesenchymal stem
neural stem cells originating from umbilical cord-derived mesenchymal stem cells improve the neurodegeneration of femoral nerve in a diabetic foot
cells in diabetic retinopathy. Scientific reports. 2017;7(1):408. ulceration rats. Neuroscience letters. 2015;597:84-9.
106. Yu B, Li XR, Zhang XM. Mesenchymal stem cell-derived extracellular vesicles 132. Luo Y, Liang F, Wan X, Liu S, Fu L, Mo J. et al. Hyaluronic Acid Facilitates
as a new therapeutic strategy for ocular diseases. World journal of stem cells. Angiogenesis of Endothelial Colony Forming Cell Combining With
2020;12(3):178-87. Mesenchymal Stem Cell via CD44/ MicroRNA-139-5p Pathway. Frontiers in
107. Fu Y, Gao X, He GH, Chen S, Gu ZH, Zhang YL. et al. Protective effects of bioengineering and biotechnology. 2022;10:794037.
umbilical cord mesenchymal stem cell exosomes in a diabetic rat model 133. Zhao L, Guo Z, Chen K, Yang W, Wan X, Zeng P. et al. Combined
through live retinal imaging. International journal of ophthalmology. Transplantation of Mesenchymal Stem Cells and Endothelial Colony-Forming
2021;14(12):1828-33. Cells Accelerates Refractory Diabetic Foot Ulcer Healing. Stem cells
108. Li W, Jin LY, Cui YB, Xie N. Human umbilical cord mesenchymal stem international. 2020;2020:8863649.
cells-derived exosomal microRNA-17-3p ameliorates inflammatory reaction 134. Hashemi SS, Mohammadi AA, Kabiri H, Hashempoor MR, Mahmoodi M,
and antioxidant injury of mice with diabetic retinopathy via targeting STAT1. Amini M. et al. The healing effect of Wharton's jelly stem cells seeded on
International immunopharmacology. 2021;90:107010. biological scaffold in chronic skin ulcers: A randomized clinical trial. Journal
109. Xu Z, Tian N, Li S, Li K, Guo H, Zhang H. et al. Extracellular vesicles secreted of cosmetic dermatology. 2019;18(6):1961-7.
from mesenchymal stem cells exert anti-apoptotic and anti-inflammatory
https://www.medsci.org
Int. J. Med. Sci. 2023, Vol. 20 1506
135. Shen WC, Liang CJ, Wu VC, Wang SH, Young GH, Lai IR. et al. Endothelial 157. Milan PB, Lotfibakhshaiesh N, Joghataie MT, Ai J, Pazouki A, Kaplan DL. et al.
progenitor cells derived from Wharton's jelly of the umbilical cord reduces Accelerated wound healing in a diabetic rat model using decellularized
ischemia-induced hind limb injury in diabetic mice by inducing HIF-1α/IL-8 dermal matrix and human umbilical cord perivascular cells. Acta
expression. Stem cells and development. 2013;22(9):1408-18. biomaterialia. 2016;45:234-46.
136. Qin HL, Zhu XH, Zhang B, Zhou L, Wang WY. Clinical Evaluation of Human 158. Montanucci P, di Pasquali C, Ferri I, Pescara T, Pennoni I, Siccu P. et al.
Umbilical Cord Mesenchymal Stem Cell Transplantation After Angioplasty Human Umbilical Cord Wharton Jelly-Derived Adult Mesenchymal Stem
for Diabetic Foot. Experimental and clinical endocrinology & diabetes : official Cells, in Biohybrid Scaffolds, for Experimental Skin Regeneration. Stem cells
journal, German Society of Endocrinology [and] German Diabetes international. 2017;2017:1472642.
Association. 2016;124(8):497-503. 159. Srifa W, Kosaric N, Amorin A, Jadi O, Park Y, Mantri S. et al.
137. Lucas T, Waisman A, Ranjan R, Roes J, Krieg T, Müller W. et al. Differential Cas9-AAV6-engineered human mesenchymal stromal cells improved
roles of macrophages in diverse phases of skin repair. Journal of immunology cutaneous wound healing in diabetic mice. Nature communications.
(Baltimore, Md : 1950). 2010;184(7):3964-77. 2020;11(1):2470.
138. Okizaki S, Ito Y, Hosono K, Oba K, Ohkubo H, Amano H. et al. Suppressed 160. Xie Y, Al-Aly Z. Risks and burdens of incident diabetes in long COVID: a
recruitment of alternatively activated macrophages reduces TGF-β1 and cohort study. The lancet Diabetes & endocrinology. 2022;10(5):311-21.
impairs wound healing in streptozotocin-induced diabetic mice. Biomedicine 161. Barron E, Bakhai C, Kar P, Weaver A, Bradley D, Ismail H. et al. Associations
& pharmacotherapy = Biomedecine & pharmacotherapie. 2015;70:317-25. of type 1 and type 2 diabetes with COVID-19-related mortality in England: a
139. Blakytny R, Jude E. The molecular biology of chronic wounds and delayed whole-population study. The lancet Diabetes & endocrinology.
healing in diabetes. Diabetic medicine : a journal of the British Diabetic 2020;8(10):813-22.
Association. 2006;23(6):594-608. 162. Roncon L, Zuin M, Rigatelli G, Zuliani G. Diabetic patients with COVID-19
140. Goren I, Kämpfer H, Podda M, Pfeilschifter J, Frank S. Leptin and wound infection are at higher risk of ICU admission and poor short-term outcome.
inflammation in diabetic ob/ob mice: differential regulation of neutrophil and Journal of clinical virology : the official publication of the Pan American
macrophage influx and a potential role for the scab as a sink for inflammatory Society for Clinical Virology. 2020;127:104354.
cells and mediators. Diabetes. 2003;52(11):2821-32. 163. Hoffmann M, Kleine-Weber H, Schroeder S, Krüger N, Herrler T, Erichsen S.
141. Mirza RE, Fang MM, Ennis WJ, Koh TJ. Blocking interleukin-1β induces a et al. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked
healing-associated wound macrophage phenotype and improves healing in by a Clinically Proven Protease Inhibitor. Cell. 2020;181(2):271-80.e8.
type 2 diabetes. Diabetes. 2013;62(7):2579-87. 164. Wysocki J, Ye M, Soler MJ, Gurley SB, Xiao HD, Bernstein KE. et al. ACE and
142. Fong CY, Tam K, Cheyyatraivendran S, Gan SU, Gauthaman K, Armugam A. ACE2 activity in diabetic mice. Diabetes. 2006;55(7):2132-9.
et al. Human Wharton's jelly stem cells and its conditioned medium enhance 165. Roca-Ho H, Riera M, Palau V, Pascual J, Soler MJ. Characterization of ACE
healing of excisional and diabetic wounds. Journal of cellular biochemistry. and ACE2 Expression within Different Organs of the NOD Mouse.
2014;115(2):290-302. International journal of molecular sciences. 2017;18(3).
143. Shrestha C, Zhao L, Chen K, He H, Mo Z. Enhanced healing of diabetic 166. Fang L, Karakiulakis G, Roth M. Are patients with hypertension and diabetes
wounds by subcutaneous administration of human umbilical cord derived mellitus at increased risk for COVID-19 infection? The Lancet Respiratory
stem cells and their conditioned media. International journal of medicine. 2020;8(4):e21.
endocrinology. 2013;2013:592454. 167. Bernardo ME, Fibbe WE. Mesenchymal stromal cells: sensors and switchers of
144. Palma MB, Luzzani C, Andrini LB, Riccillo F, Buero G, Pelinski P. et al. Wound inflammation. Cell stem cell. 2013;13(4):392-402.
Healing by Allogeneic Transplantation of Specific Subpopulation From 168. Hernandez JJ, Beaty DE, Fruhwirth LL, Lopes Chaves AP, Riordan NH.
Human Umbilical Cord Mesenchymal Stem Cells. Cell transplantation. Dodging COVID-19 infection: low expression and localization of ACE2 and
2021;30:963689721993774. TMPRSS2 in multiple donor-derived lines of human umbilical cord-derived
145. Huang J, Wu S, Wu M, Zeng Q, Wang X, Wang H. Efficacy of the therapy of mesenchymal stem cells. Journal of translational medicine. 2021;19(1):149.
5-aminolevulinic acid photodynamic therapy combined with human umbilical 169. Lanzoni G, Linetsky E, Correa D, Messinger Cayetano S, Alvarez RA,
cord mesenchymal stem cells on methicillin-resistant Staphylococcus Kouroupis D. et al. Umbilical cord mesenchymal stem cells for COVID-19
aureus-infected wound in a diabetic mouse model. Photodiagnosis and acute respiratory distress syndrome: A double-blind, phase 1/2a, randomized
photodynamic therapy. 2021;36:102480. controlled trial. Stem cells translational medicine. 2021;10(5):660-73.
146. Zhang Y, Zhang P, Gao X, Chang L, Chen Z, Mei X. Preparation of exosomes 170. Khanh VC, Fukushige M, Chang YH, Hoang NN, Yamashita T,
encapsulated nanohydrogel for accelerating wound healing of diabetic rats by Obata-Yasuoka M. et al. Wharton's Jelly Mesenchymal Stem Cell-Derived
promoting angiogenesis. Materials science & engineering C, Materials for Extracellular Vesicles Reduce SARS-CoV2-Induced Inflammatory Cytokines
biological applications. 2021;120:111671. Under High Glucose and Uremic Toxin Conditions. Stem cells and
147. Yan C, Xv Y, Lin Z, Endo Y, Xue H, Hu Y. et al. Human Umbilical Cord development. 2021;30(15):758-72.
Mesenchymal Stem Cell-Derived Exosomes Accelerate Diabetic Wound 171. Hashemian SR, Aliannejad R, Zarrabi M, Soleimani M, Vosough M, Hosseini
Healing via Ameliorating Oxidative Stress and Promoting Angiogenesis. SE. et al. Mesenchymal stem cells derived from perinatal tissues for treatment
Frontiers in bioengineering and biotechnology. 2022;10:829868. of critically ill COVID-19-induced ARDS patients: a case series. Stem cell
148. Wei Q, Wang Y, Ma K, Li Q, Li B, Hu W. et al. Extracellular Vesicles from research & therapy. 2021;12(1):91.
Human Umbilical Cord Mesenchymal Stem Cells Facilitate Diabetic Wound 172. Tao J, Nie Y, Wu H, Cheng L, Qiu Y, Fu J. et al. Umbilical cord blood-derived
Healing Through MiR-17-5p-mediated Enhancement of Angiogenesis. Stem mesenchymal stem cells in treating a critically ill COVID-19 patient. Journal of
cell reviews and reports. 2022;18(3):1025-40. infection in developing countries. 2020;14(10):1138-45.
149. Zhang S, Chen L, Zhang G, Zhang B. Umbilical cord-matrix stem cells induce 173. Ma W, Wu JH, Wang Q, Lemaitre RN, Mukamal KJ, Djoussé L. et al.
the functional restoration of vascular endothelial cells and enhance skin Prospective association of fatty acids in the de novo lipogenesis pathway with
wound healing in diabetic mice via the polarized macrophages. Stem cell risk of type 2 diabetes: the Cardiovascular Health Study. The American
research & therapy. 2020;11(1):39. journal of clinical nutrition. 2015;101(1):153-63.
150. Jung JA, Yoon YD, Lee HW, Kang SR, Han SK. Comparison of human 174. Boland LK, Burand AJ, Boyt DT, Dobroski H, Di L, Liszewski JN. et al. Nature
umbilical cord blood-derived mesenchymal stem cells with healthy fibroblasts vs. Nurture: Defining the Effects of Mesenchymal Stromal Cell Isolation and
on wound-healing activity of diabetic fibroblasts. International wound Culture Conditions on Resiliency to Palmitate Challenge. Frontiers in
journal. 2018;15(1):133-9. immunology. 2019;10:1080.
151. You HJ, Namgoong S, Han SK, Jeong SH, Dhong ES, Kim WK. Wound-healing 175. El-Hossary N, Hassanein H, El-Ghareeb AW, Issa H. Intravenous vs
potential of human umbilical cord blood-derived mesenchymal stromal cells intraperitoneal transplantation of umbilical cord mesenchymal stem cells from
in vitro--a pilot study. Cytotherapy. 2015;17(11):1506-13. Wharton's jelly in the treatment of streptozotocin-induced diabetic rats.
152. Moon KC, Lee JS, Han SK, Lee HW, Dhong ES. Effects of human umbilical Diabetes research and clinical practice. 2016;121:102-11.
cord blood-derived mesenchymal stromal cells and dermal fibroblasts on 176. Burand AJ, Jr., Di L, Boland LK, Boyt DT, Schrodt MV, Santillan DA. et al.
diabetic wound healing. Cytotherapy. 2017;19(7):821-8. Aggregation of Human Mesenchymal Stromal Cells Eliminates Their Ability
153. Nilforoushzadeh MA, Raoofi A, Afzali H, Gholami O, Zare S, Nasiry D. et al. to Suppress Human T Cells. Frontiers in immunology. 2020;11:143.
Promotion of cutaneous diabetic wound healing by subcutaneous 177. Yang XF, Chen T, Ren LW, Yang L, Qi H, Li FR. Immunogenicity of
administration of Wharton's jelly mesenchymal stem cells derived from insulin-producing cells derived from human umbilical cord mesenchymal
umbilical cord. Archives of dermatological research. 2023 Mar;315(2):147-159. stem cells. Experimental and therapeutic medicine. 2017;13(4):1456-64.
154. Han Y, Sun T, Han Y, Lin L, Liu C, Liu J. et al. Human umbilical cord 178. Ngoc PK, Phuc PV, Nhung TH, Thuy DT, Nguyet NT. Improving the efficacy
mesenchymal stem cells implantation accelerates cutaneous wound healing in of type 1 diabetes therapy by transplantation of immunoisolated
diabetic rats via the Wnt signaling pathway. European journal of medical insulin-producing cells. Human cell. 2011;24(2):86-95.
research. 2019;24(1):10. 179. Teotia RS, Kadam S, Singh AK, Verma SK, Bahulekar A, Kanetkar S. et al. Islet
155. Yue C, Guo Z, Luo Y, Yuan J, Wan X, Mo Z. c-Jun Overexpression Accelerates encapsulated implantable composite hollow fiber membrane based device: A
Wound Healing in Diabetic Rats by Human Umbilical Cord-Derived bioartificial pancreas. Materials science & engineering C, Materials for
Mesenchymal Stem Cells. Stem cells international. 2020;2020:7430968. biological applications. 2017;77:857-66.
156. Yang J, Chen Z, Pan D, Li H, Shen J. Umbilical Cord-Derived Mesenchymal 180. Li X, Wei Z, Wu L, Lv H, Zhang Y, Li J. et al. Efficacy of
Stem Cell-Derived Exosomes Combined Pluronic F127 Hydrogel Promote Fe(3)O(4)@polydopamine nanoparticle-labeled human umbilical cord
Chronic Diabetic Wound Healing and Complete Skin Regeneration. Wharton's jelly-derived mesenchymal stem cells in the treatment of
International journal of nanomedicine. 2020;15:5911-26. streptozotocin-induced diabetes in rats. Biomaterials science.
2020;8(19):5362-75.
https://www.medsci.org
Int. J. Med. Sci. 2023, Vol. 20 1507
181. Montanucci P, Pescara T, Greco A, Leonardi G, Marini L, Basta G. et al.
Co-microencapsulation of human umbilical cord-derived mesenchymal stem
and pancreatic islet-derived insulin producing cells in experimental type 1
diabetes. Diabetes/metabolism research and reviews. 2021;37(2):e3372.
182. Bari S, Chu PP, Lim A, Fan X, Gay FP, Bunte RM. et al. Protective role of
functionalized single walled carbon nanotubes enhance ex vivo expansion of
hematopoietic stem and progenitor cells in human umbilical cord blood.
Nanomedicine : nanotechnology, biology, and medicine. 2013;9(8):1304-16.
183. Fan X, Gay FP, Lim FW, Ang JM, Chu PP, Bari S. et al. Low-dose insulin-like
growth factor binding proteins 1 and 2 and angiopoietin-like protein 3
coordinately stimulate ex vivo expansion of human umbilical cord blood
hematopoietic stem cells as assayed in NOD/SCID gamma null mice. Stem
cell research & therapy. 2014;5(3):71.
https://www.medsci.org