CC BY 4.0 · Eur J Dent 2024; 18(03): 712-742
DOI: 10.1055/s-0043-1776315
Review Article

Angiogenic Potential of Various Oral Cavity–Derived Mesenchymal Stem Cells and Cell-Derived Secretome: A Systematic Review and Meta-Analysis

1   Department of Oral Pathology and Microbiology, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune, India
,
1   Department of Oral Pathology and Microbiology, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune, India
,
2   Department of Pediatric and Preventive Dentistry, Bharati Vidyapeeth (Deemed to be) University Dental College and Hospital, Navi Mumbai, India
,
3   Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune, India
,
3   Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune, India
,
3   Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune, India
,
4   Department of Oral Pathology and Microbiology, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune, India
,
5   Department of Prosthodontics, M.A. Rangoonwala College of Dental Sciences and Research Centre, Pune, India
,
6   Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune, India
,
7   Department of Oral and Maxillofacial Surgery, Dr. D. Y. Patil Dental College, and Hospital, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune, India
› Author Affiliations
 

Abstract

Recent evidence suggests the immense potential of human mesenchymal stem cell (hMSC) secretome conditioned medium-mediated augmentation of angiogenesis. However, angiogenesis potential varies from source and origin. The hMSCs derived from the oral cavity share an exceptional quality due to their origin from a hypoxic environment. Our systematic review aimed to compare the mesenchymal stem cells (MSCs) derived from various oral cavity sources and cell-derived secretomes, and evaluate their angiogenic potential. A literature search was conducted using PubMed and Scopus from January 2000 to September 2020. Source-wise outcomes were systematically analyzed using in vitro, in vivo, and in ovo studies, emphasizing endothelial cell migration, tube formation, and blood vessel formation. Ninety-four studies were included in the systematic review, out of which 4 studies were subsequently included in the meta-analysis. Prominent growth factors and other bioactive components implicated in improving angiogenesis were included in the respective studies. The findings suggest that oral tissues are a rich source of hMSCs. The meta-analysis revealed a positive correlation between dental pulp–derived MSCs (DPMSCs) and stem cells derived from apical papilla (SCAP) compared to human umbilical cord–derived endothelial cell lines as a control. It shows a statistically significant positive correlation between the co-culture of human umbilical vein endothelial cells (HUVECs) and DPMSCs with tubule length formation and total branching points. Our meta-analysis revealed that oral-derived MSCs (dental pulp stem cells and SCAP) carry a better angiogenic potential in vitro than endothelial cell lines alone. The reviewed literature illustrates that oral cavity–derived MSCs (OC-MSCs) increased angiogenesis. The present literature reveals a dearth of investigations involving sources other than dental pulp. Even though OC-MSCs have revealed more significant potential than other MSCs, more comprehensive, target-oriented interinstitutional prospective studies are warranted to determine whether oral cavity–derived stem cells are the most excellent sources of significant angiogenic potential.


#

Introduction

Oral cavity–derived dental pulp stem cells (DPSCs) have gained attention due to their potential use in regenerative medicine. These stem cells are known for their unique characteristics that make them distinct from other stem cell sources. Some exceptional criteria of oral cavity–derived DPSCs are their mesenchymal stem cell (MSC) characteristics, ease of accessibility, multilineage differentiation, regenerative capacity with high angiogenic potential, and immunomodulatory properties with low immunogenicity. Despite their potential advantages, using oral cavity–derived stem cells for oral cancer treatment and reconstruction poses several challenges. Oral cancer creates a hostile tumor microenvironment characterized by inflammation, hypoxia, and immune suppression. Stem cells may face difficulty surviving and exerting their regenerative properties in such an environment. There is a risk that the harvested stem cell population could be contaminated with cancer cells, which can lead to cancer recurrence if transplanted back into the patient. Moreover, oral carcinoma contains a population of neoplastic cells with aggressive stem cells that are difficult to distinguish from healthy cells. Angiogenesis or neovascularization is a dynamic process involving new blood vessels that form from existing blood vessels.[1] Oral cavity stem cells secrete various angiogenic factors, including vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), platelet-derived growth factor (PDGF), and others. These factors attract endothelial cells and support the formation of new capillaries.

Oral cavity stem cells can trigger angiogenesis when introduced into a tissue requiring regeneration or healing. It is crucial during development, along with various physiological and pathological processes.[2] Angiogenesis occurs lifelong, starting in the uterus and continuing into old age. Furthermore, capillaries are required to exchange nutrients and metabolites in all tissues.[3] Angiogenesis is paramount concerning wound healing due to its critical role in growing a new capillary network from the granulation tissue, which plays a pivotal role in chronic inflammation.[3] Wound healing is a complex procedure involving overlapping events, including inflammatory, proliferative, and remodeling phases. Many growth factors and cytokines participate in the proliferative phase, of which angiogenic growth factors hold a prime role.[4] Revascularization is regulated by a complex interaction between various growth factors, including but not limited to VEGF, FGF, angiopoietins (ANG), PDGF, transforming growth factor-α (TGF-α), and transforming growth factor-β (TGF-β).[5] Each factor plays a separate role in inducing, initiating, and amplifying cell proliferation, cell migration, stabilization, wound healing, inflammation, and suppression of angiogenesis.[1] [6] Several growth factors like VEGF, FGF-2, and PDGF have been used clinically to augment angiogenesis for various therapeutic applications. However, lack of spatiotemporal control over the release of these proangiogenic proteins has led to numerous complications, including leaky vasculature. Cell-based therapies are evolving therapeutic options for deranged angiogenesis.[7]

MSCs derived from human placental tissue, bone marrow, or umbilical cord tissues provide a novel strategy for the induction of angiogenesis. Various studies have demonstrated the ability of MSCs to differentiate into endothelial cells and provide vascular stability. In addition, MSCs secrete an extended milieu of growth factors, cytokines, extracellular vesicles (EVs), and messenger ribonucleic acids (mRNAs) implicated in a wide range of biological processes. Interestingly, “secret factors” (secretomes) from MSCs promote angiogenesis and amend wound healing in virtue of potent paracrine signaling, yielding proangiogenic factors.[5] Although hMSCs isolated from various sources have exhibited proangiogenic potential, knowledge about the ideal source (cells or secretomes, source-wise potential, and ease of sample collection) remains obscure. Oral tissues originate from mesenchymal and ectodermal germ layers that add to their value, making them the ideal source for isolation and therapeutic applications. Stem cells are influenced by their in vivo environment, which projects through their therapeutic properties.[8] The stem cell niche includes cellular and extracellular matrix components, tissue location, innervation, and blood supply. The oral cavity is highly vascularized and yields better-quality stem cells with potent angiogenic potential. Rapid wound healing in the oral cavity can explain its unique potential. Their high proliferation and unique secretory profile can be attributed to their hypoxic condition. Oral cavity–derived cells are multipotent; primitive oral tissues such as dental follicles harbor oral cavity–derived MSCs (OC-MSCs). Therefore, MSCs isolated from various sources from the oral cavity comprise a powerful weapon to battle numerous diseases.[9]

In recent decades, stem cell proliferation from various adult tissues has been a provoking tool in advanced sciences. Previous studies have revealed the role of dental pulp–derived MSCs (DPMSCs) and stem cells from human exfoliated deciduous teeth (SHED) in enhancing the cascade of angiogenesis. Our systematic review aimed to compare OC-MSCs and cell-derived secretomes and evaluate their angiogenic potential. The subsequent meta-analysis with compatible data analyses whether OC-MSCs (DPSC and stem cells derived from apical papilla [SCAP]) carry a better angiogenic potential in vitro than endothelial cell lines alone. Extensive collaborative research is required to conclude which oral-derived stem cells have the best angiogenic potential. This systematic review focuses on the potential of MSCs and their secretomes derived from various oral tissues such as gingival tissue, dental pulp, periodontal ligament (PDL), mandibular bone, and buccal fat, with particular emphasis on angiogenesis.


#

Methods

This study was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. The research proposal has been registered in PROSPERO (registration no: CRD42021282497).

Based on the PRISMA criteria, the research question for this review was framed in the PICO format as the following: Which is the best oral source of MSCs for augmenting angiogenesis at the implanted site?

The terms used to identify studies based on the elements of the PICO format were as follows:

  • Population: in vitro studies, in vivo studies, and ex vivo studies.

  • Intervention: OC-MSCs.

  • Comparison: between various OC-MSCs.

  • Outcome: angiogenesis at the desired site of implantation.

The inclusion criteria of the study were the following:

  • Articles published in the English language.

  • Studies relevant to the topic published from January 1, 2000 to March 2023.

  • Studies showing in vitro, in vivo, and in ovo results for angiogenesis of OC-MSCs.

  • Studies having well-defined information regarding the angiogenic potential of OC-MSCs.

The exclusion criteria of the review included the following:

  • Abstracts.

  • Reviews.

  • Letter to the editor.

  • Editorials.

  • Case reports.

  • Short communication.

  • Commentaries.

  • Articles in languages other than English.

Systematic computer searches were performed on two electronic databases: PubMed and Scopus. The following keyword combinations were used to search articles:

  • “Dental stem cells AND Angiogenesis AND conditioned media.”

  • “Dental stem cells AND Angiogenic potential AND conditioned media.”

  • “Dental stem cells AND Angiogenesis.”

  • “Dental stem cells AND Angiogenic potential.”

Along with the electronic search, a hand search was also performed to find the missed articles. Articles published between January 1, 2000 and March 1, 2023 were included in the survey. Two reviewers (M.S. and S.K.) independently evaluated the titles and abstracts of the retrieved publications pertaining to the covered research topic during the initial screening. If material relevant to the inclusion criteria was provided in the abstract, or if the title was relevant but the abstract was unavailable, a full-text report was acquired. The complete text of the articles was then screened to find those that matched the inclusion criteria. If the work appeared to meet the inclusion criteria, the authors were contacted to seek further information. Articles with full-text reports only were evaluated in this systematic review. Studies that only published abstracts were removed because evidence revealed differences between data given in abstracts and those supplied in the final published complete report. Two review authors (M.S. and S.K.) separately collected data using a specifically designed data extraction sheet ([Table 1]). A third (S.D.) and a fourth (Y.M.) reviewer handled any disagreements about the inclusion of publications or data extraction.

Table 1

Tabular representation of qualitative data obtained from literature search for included studies

Sl. no.

Study

Source of stem cells

Type of study

Model used

Factors assessed for angiogenesis

Method used for analysis

Use of stem cells/conditioned media

Use of preconditioning (yes/no)

Use of co-culture with MSCs (yes/no)

Results obtained

Dental pulp–derived mesenchymal stem cells (DPMSCs)

1.

Li et al[10]

Dental pulp

In vivo

Mice

VEGFR1, VEGFR2, VE-cadherin, ETV2, and CD31

Real-time polymerase chain reaction (RT-PCR)

Cells

ETV2 transfected

Human umbilical vein endothelial cell (HUVEC)

Dental pulp stem cell (DPSCs) proved as potential candidates for clinical applications in therapeutic tissue engineering

2.

Boreak et al[11]

Dental pulp

In ovo

Yolk sac membrane (YSM)

VEGFA, FGF-2, CXCL8, VEGF, and angiopoietin-2

Enzymelinked immunosorbent assay (ELISA) and RT-PCR

Conditioned media

Metformin, cisplatin (negative control)

L-arginine (positive control)

No

Metformin treated conditioned media derived from DPSCs enhanced the level of angiogenic activity in the YSM

3.

Li et al[12]

Dental pulp

In vivo

Rats

Angiogenin, basic fibroblast growth factor (bFGF), hepatocyte growth factor (HGF), HIF-1α, interleukin-8 (IL-8), monocyte chemotactic protein 1 (MCP-1), platelet-derived growth factor (PDGF), and vascular endothelial growth factor (VEGF)

RT-PCR

Cells

No

No

Stem cells from the dental pulp provided greater therapeutic effects compared to stem cells from the umbilical cord

4.

Li et al[13]

Dental pulp

In vivo

Rats

VEGF, VEGFR-2 (Flk1)

RT-PCR and ELISA

Cells

Nell-1

HUVEC

Nell-1 could promote endothelial vessel formation and enhance the angiogenic factor expression when treated over the DPSCs or HUVECs

5.

Alghutaimel et al[14]

Dental pulp

In vivo

Mice

VEGF-A, FGF-2

RT-PCR

Cells

Decellularized dental pulp (DDP) matrix of bovine origin treated the DPSCs

No

DDP seeded along with the DPSCs provided greater angiogenic efficiency that singularly seeded the DDP

6.

Zhou et al[15]

Dental pulp

In vitro

HUVECs

VEGF

RT-PCR

Cells

Transfection of miR-378a. hedgehog/Gli1 signaling inhibition

HUVECs

Extracellular vesicles derived from the DPSC transfected with miR-378a could enhance angiogenic proliferation in vitro

7.

Huang et al[16]

Dental pulp

In vitro

HUVECs

VEGF and kinase-insert domain-containing receptor (KDR)

RT-PCR

Cells

Lipopolysaccharide (LPS)

HUVECs

Inflammatory stimulation

8.

Afami et al[17]

Dental pulp

In vitro

Microbes

Angiogenin, EGF, FGF, PDGF, INF-gamma, VEGF, insulinlike growth factor (IGF), and angiopoietin

Heat map

Both

(Naphthalene-2-ly)-acetyl-diphenylalanine-dilysine-OH (NapFFεKεK-OH)

Hydrogel

Increased vascular components

9.

Liao et al[18]

Dental pulp

In vivo

Mice

VEGF and AngII

qRT-PCR analysis and immunofluorescence staining

Cells

No

No

Enhanced wound healing

10.

He et al[19]

Dental pulp

In vitro

C. albicans biofilms

Hyphal wall protein1 (hwp1), agglutininlike sequence protein 3 (als3) and cell surface hydrophobicity (csh1)

RT-PCR

Cells

Norspermidine (NSPD)

GelMA hydrogels

NSPD did not directly influence the angiogenic properties of the DPSCs

11.

Guo et al[20]

Dental pulp

In vivo

Rat

Angiogenin, EGF, bFGF, and HGF

Not mentioned

Cells

No

Human adipose microvascular endothelial cells (HAMECs)

Co-culture of the DPSC with the HAMECs yielded denser vascular bundles compared to endothelial cells alone

12.

Luzuriaga et al[21]

Dental pulp

In vitro

Mouse liver sinusoidal endothelial cells (mLSECs)

VEGF

Flow cytometry

Cells

No

No

Use of the DPSC-enhanced prevascularized engraftments improves cell–graft integration compared to nonvascularized grafts

13.

Merckx et al[22]

Dental pulp

In ovo

Chorioallantoic membrane (CAM) of chicken embryos

Angiogenin, angiopoietin-1 (Angpt-1), HGF, insulinlike growth factor-binding proteins (IGFBPs), monocyte chemoattractant protein-1 (MCP-1), urokinase plasminogen activator (uPA), and VEGF

Transmission electron microscopy, high-resolution flow cytometry, and ELISA

Both

No

Co-culture with bone marrow–derived MSCs (BM-MSCs)

Positive paracrine effects on endothelial cell migration and in ovo blood vessel formation, with a stronger potential for BM-MSCs was found

14.

Caseiro et al[23]

Dental pulp

In vivo

Rats

Angiopoietin-2 (Ang), EGF, endothelin-1 (EDN1), fibroblast growth factor 1 and 2 (FGF-1 and FGF-2), PDGF-AA and PDGF-AB/BB, transforming growth factor alpha (TGFα), transforming growth factor beta 1, 2, and 3 (TGF-β1, -2, and -3), tumor necrosis factor alpha (TNFα), TNFβ, VEGF-A, VEGF-C, and VEGF-D

PCR

Both

No

Co-culturing was done with umbilical cord–derived MSCs

UC-MSCs provide a wider variety and greater concentration of relevant growth factors and cytokines

15.

Makino et al[24]

Dental pulp

In vivo

Rats

TNF-α, VEGF, and bFGF

Immunohistological staining

Both

No

No

Increased capillary formation achieved

16.

Chen et al[25]

Dental pulp

In vitro

Endothelial cell line

VEGF, PDGF, SDF-1, and GAPDH

IHC and PCR

Cells

No

No

VEGF expression was higher in pulp tissue from teeth with deep caries (cDPMSCs) than in normal tissue

17.

Li et al[26]

Dental pulp

In vitro

Endothelial cell line

KDR and CD31

Immunofluorescence analysis and RT-PCR

Cells

No

Human decellularized dental pulp matrix (hDDPM)

Increased proliferation of blood vessel-like structures was evident

18.

Wang et al[27]

Dental pulp

In vitro

Endothelial cell line

VEGF

ELISA, two-photon laser microscopy

Cells

No

HUVECs

Extracellular vesicles from the DPMSCs can promote angiogenesis in an injectable hydrogel in vitro

19.

Zhou and Sun[28]

Dental pulp

In vitro

Endothelial cell line under hypoxic conditions

VEGF, FGF, vWF, VEGFR2, VE-cad, HIF-1α, and CD31

PCR

Cells

No

No

Hypoxic conditions enhanced the tube formation of the DPMSCs in vitro

20.

Qu et al[29]

Dental pulp

In vitro

HUVECs

Angiopoietin-1, VEGFA, and ribosomal protein L13a (RPL13a)

PCR and ELISA

Both

No

No

DPMSCs derived from conditioned medium (CM) could enhance capillary tube formation

21.

Zhu et al[30]

Dental pulp

In vivo

Mice

VEGF and SDF-1α

PCR and ELISA

Both

No

No

Enhanced expression of VEGF and SDF-1α was observed

22.

Li et al[31]

Dental pulp

In vitro

Endothelial cell line

VEGF, FGF, ANG-1, and PDGFA

RT-PCR and immunofluorescence

Cells

IGFBP5

No

IGFBP5 overexpression enhanced the expressions of angiogenic differentiation markers

23.

Lu et al[32]

Dental pulp

In vitro

Endothelial cell line

p-AKT and cyclin D1

RT-PCR and Western blotting

Cells

VEGF and IGF-1

No

Combined treatment with VEGF and IGF-1 provided a synergistic effect on the angiogenic potential of DPMSCs derived from carious teeth

24.

Youssef et al[33]

Dental pulp

In vitro

Endothelial cell line

VEGF

PCR, flow cytometry

Cells

Mineral trioxide aggregate (MTA), calcium hydroxide (Ca [OH]2, Biodentine (BD) and Emdogain

No

The treatment of MTA-enhanced VEGF expression, Ca (OH)2, BD, and Emdogain

25.

Rapino et al[34]

Dental pulp

In vitro

Endothelial cell line

EDN1, VEGF, IL–6, and PGE2

ELISA

Cells

Chitlac-coated BisGMA/triethylene glycol dimethacrylate (TEGDMA) methacrylic thermosets

No

The addition of Chitlac-coated BisGMA/TEGDMA methacrylic thermosets resulted in tubules with an increased diameter and improved the differentiation of angiogenic cell types

26.

Dubey et al[35]

Dental pulp

In vitro

Endothelial cell line

VEGF

Light and fluorescence microscopy

Cells

Clindamycin (CLIN) and minocycline (MINO)

HUVECs

There was enhanced cell proliferation and capillary tube formation

27.

Delle Monache et al[36]

Dental pulp

In vitro

Endothelial cell line

FGF, VEGF, and EGF

Immunofluorescence, and Western blotting

Cells

Complete endothelial medium 2 (EGM-2)

HUVECs

EGM-2-treated DPMSCs formed tubelike structures that were more stabilized compared to HUVECs alone

28.

Gong et al[37]

Dental pulp

In vitro

Endothelial cell line

VEGF

Immunofluorescence microscopy, PCR, and ELISA

Cells

EphrinB2-Fc or EphB4-Fc

HUVECs

EphrinB2-Fc or EphB4-Fc enhanced the DPMSCs to form blood vessels with increased secretion of VEGF

29.

Schertl et al[38]

Dental pulp

In vitro

Endothelial cell line

PECAM1, VEGF-A, and KDR

Flow cytometry, and qRT-PCR analysis

Cells

TEGDMA

No

Treatment with 0.25 mM of TEGDMA downregulated angiogenic factor expression, while at 0.1 mM concentration angiogenesis was not affected

30.

Luzuriaga et al[39]

Dental pulp

In vivo

Mouse

VEGF

PCR, flow cytometry, and Western blotting

Cells

No

No

Dental pulp–derived cells contributed to the generation of neovasculature in brain tissue

31.

Zou et al[40]

Dental pulp

In vitro

Endothelial cell line

VEGF, HIF-1α, ANG1, and ANGPTL4

ELISA

Cells

Sema 4D/plexin B1

No

Signaling through sema 4D/plexin B1-induced endothelial differentiation of the DPMSCs

32.

Bindal et al[6]

Dental pulp

In vitro

Endothelial cell line

FGF, VEGF-A, HGF, PDGF-BB, MCP-1, and CCL5

RT-qPCR array

Cells

LPS, human platelet lysate (HPL), platelet-rich plasma

No

20% HPL has been shown to provide the most optimal environment to induce proangiogenic factors in inflammatory DPMSCs

33.

Jin et al[41]

Dental pulp

In vitro

Endothelial cell line

VEGF, FGF, PDGF, TGF-β

RT-PCR and immunofluorescence

Cells

Concentrated growth factor (CGF) scaffold

HUVECs

At low doses, CGF could potentially stimulate endothelial cell proliferation and migration

34.

Gharaei et al[42]

Dental pulp

In vitro

HUVEC line

VEGF, IGF-1, SDF-1, IGFBP-2,3, MMP-9, TIMP-1, and Ang-1

ELISA, RT-PCR, and protein profiling array

Both

No

No

CM released from hDPMSCs can trigger pronounced angiogenic effects

35.

Dou et al[43]

Dental pulp

In vitro

Endothelial cell line

VEGFA, HIF-1A, KDR(VEGFR2), TGFβ1, BMP-2, bFGF, HGF, TNF-α, Runx-2, and Notch-1

PCR, flow cytometry, and ELISA

Cells

Hypoxic conditions

No

Hypoxia could promote angiogenesis of the DPMSCs graft via the HIF-1ɑ signaling pathway

36.

Aksel et al[44]

Dental pulp

In vitro

Endothelial cell line

VEGF

ELISA and PCR

Cells

Fibrin gel integrated demineralized dentin matrix

No

Increased angiogenic marker expression

37.

Lambrichts et al[45]

Dental pulp

In ovo and in vivo

Chorioallantoic membrane, mice

VEGF, angiogenin, dipeptidyl peptidase IV, angiopoietin-1, EDN1, IGFBP-3, IL-8, urokinase-type plasminogen activator, MCP-1

Histopathologic staining

Both

No

No

hDPMSCs significantly augmented blood vessel growth in this ovo model for angiogenesis; also, pulp vascularization was obtained in a transplanted scaffold in the immune-compromised mice model

38.

Silva et al[46]

Dental pulp

In vivo

Mice

VEGF, VEGFR2, and IL-8

ELISA

Cells

Lipoprotein receptor–related protein 6 (LRP6) and Frizzled6, recombinant human Wnt1 (rhWnt1), and recombinant human VEGF165 (rhVEGF165)

No

Lipoprotein receptor–related protein 6 silenced DPMSCs downregulated VEGF expression also showed fewer blood vessel formation in the mice model

39.

Aksel and Huang[47]

Dental pulp

In vitro

Endothelial cell line

von Willebrand factor (vWF)

Immunofluorescence

Cells

Endothelial growth medium-2 (EGM-2)

No

EGM-2-induced cells showed improved vessel formation compared to noninduced cells

40.

Zou et al[48]

Dental pulp

In vitro

Endothelial cell line

VEGF

ELISA and PCR

Cells

Sema4D/plexin B1

HUVECs

Sema4D/plexinB1 signaling exerts profound effects on enhancing VEGF secretion and angiogenesis

41.

Nam et al[49]

Dental pulp

In vivo

Mice

VEGF, α-smooth muscle actin (α-SMA), PDGF receptor β (PDGFRβ), and CD146

Immunofluorescent staining

Cells

No

HUVECs

42.

Lee et al[50]

Dental pulp

In vitro

HUVECs

VEGF, FGF-2, VEGFRs, PECAM-1, and VE-cadherin

PCR

Both

Nanocomposite cements

No

The conditioning with nanocomposite cements-hDPMSC-CM showed the highest tubular number of HUVECs

43.

Lee et al[51]

Dental pulp

In vitro

HUVECs

VEGF, PDGF, FGF-2, platelet endothelial cell adhesion molecule 1 (PECAM-1), and VE-cadherin

PCR

Both

Baicalein

No

Baicalein conditioning increased capillarylike tube formation significantly

44.

Spina et al[52]

Dental pulp

In vitro

Collagen scaffolds

VEGF and PDGFA

PCR and IHC

Cells

New Zealand Foetal Bovine Serum

No

Expression of VEGF and PDGFA. hDPMSCs cultured in NZ-FBS were found to produce higher mRNA levels of the said angiogenic factors

45.

Kuang et al[53]

Dental pulp

In vivo

Mice

VEGF and HIF-1α

PCR

Cells

Hypoxic conditions

No

After 4 weeks, the hypoxia group significantly enhanced angiogenesis inside the pulp chamber

46.

Shen et al[54]

Dental pulp

In vivo

Mice

VEGF, SDF-1, MCP-1, PDGF-BB, IGF-1, TGF-β, and bFGF

IHC, laser Doppler flowmetry

Both

No

No

DP-CM was shown to significantly improve the recovery of persistent blood flow in the ischemic hindlimb of mice

47.

Dissanayaka et al[55]

Dental pulp

In vivo

Mice

VEGF

ELISA

Cells

No

HUVECs

The extracellular matrix produced by the DPMSCs promoted the stabilization and remodeling of capillarylike structures formed by the HUVECs

48.

Boyle et al[56]

Dental pulp

In vitro

HUVECs

VEGF

PCR, flow cytometry

Cells

TNF alpha

No

TNF alpha increased the angiogenesis of DPMSCs

49.

Liu et al[57]

Dental pulp

In vitro

HUVECs

VEGF, kinase insert domain receptor (KDR), and FGF

Western blotting and RT-PCR

Lentiviral vector-transfected cells

MiR-424

No

Inhibition of miR-424 function promoted endothelial cell differentiation of hDPMSCs, whereas miR-424 overexpression inhibited their angiogenic potential

50.

Bronckaers et al[58]

Dental pulp

In ovo

Human microvascular endothelial cell line 1 (HMEC-1), chicken chorioallantoic membrane, mouse brain endothelial cells (MBECs)

VEGF, IL-8, MCP-1, and FGF-2

ELISA and RT-PCR

Both

No

No

An increased number of capillary formations was evident

51.

Janebodin et al[59]

Dental pulp

In vivo

Mice

VEGF

PCR

Cells

No

BM-MSCs

DPMSCs' ability to induce vessel formation was more efficient than BMSCs

52.

Ishizaka et al[60]

Dental pulp

In vivo

Mice

Granulocyte monocyte colony-stimulating factor (GM-CSF), matrix metalloproteinase-3 (MMP-3), and VEGF-A

Flow cytometry

Both

No

Bone marrow, adipose tissue MSCs

DPMSCs have more significant potential for angiogenesis

53.

Dissanayaka et al[61]

Dental pulp

In vitro

HUVECs

CD117, VEGF, CD34, and Flk-1

PCR

Cells

No

Endothelial cells

Matrigel assay showed that the addition of DPMSCs stabilized preexisting vessel-like structures formed by endothelial cells and increased their longevity

54.

Iohara et al[62]

Dental pulp

In vivo

Mice

VEGF, MMP, CSF, CXCR4, and SDF1/CXCL12

PCR

Cells

No

No

It improved limb ischemia in the hindlimb of the mice model

Stem cells from human exfoliated deciduous teeth (SHED)

1.

Wu et al[63]

SHED

In vivo

Mice

VEGFA, PDGFA, and angiopoietin

RT-PCR

Cells

No

HUVEC and SHED exosomes

SHED exosomes provide expanded possibilities to enhance angiogenesis and pulp regeneration

2.

Han et al[64]

SHED

In vivo

Mice

VEGF

ELISA

Both

Transfection of premade siRNA for HIF-1 alpha signal silencing

HUVECs

HIF-1 alpha signaling along with VEGF has a potent role for the use of SHED in regenerative medicine

3.

Zaw et al[65]

SHED

In vitro

HUVECs

Bcl-2, NF-κB1, VEGFA, CXCL8, and CXCR1

ELISA, PCR, and flow cytometry

Cells

NF-κB decoy oligodeoxynucleotides (ODNs) or scramble (control)

Human dermal microvascular endothelial cells (HDMECs)

Increased expression of angiogenic factors was observed with co-culture

4.

Atlas et al[66]

SHED

In vivo

Mice

VEGF, HGF, and PDGF-BB

Not mentioned

Cells

No

Endothelial cells

SHED takes part in the prevascularization process to further cause maturation of the vasculature

5.

Guo et al[67]

SHED

In vivo

Minipigs

HIF-1a and VEGF

RT-PCR

Cells

No

Regenerated dental pulp stem cells and SHED together (R-SHED), HUVEC

The tube forming parameters on a Matrigel showed highest results for R-SHED. Likewise, the expression of angiogenic markers were higher in R-SHED group compared to the controls

6.

Wang et al[4]

SHED

In vitro

HUVECs

VEGF, VEGFR2 CD31 and DLL4

PCR and ELISA

Cells

Treatment with shear stress.

No

Shear stress–induced arterial endothelial differentiation of SHED and VEGF-DLL4/Notch-EphrinB2 signaling was involved in this process

7.

Gong et al[68]

SHED

In vitro

HUVECs

VEGF, FGF beta, and hEGF

IHC and PCR

Cells

No

HUVECs and decellularized matrix

Endothelial-induced SHED provided better angiogenesis

8.

Kim et al[69]

SHED

In vivo

Mice

PCR and IHC

Cells

No

HUVECs

Co-culture of HUVECs and SHED could provide enhanced angiogenesis in vivo

9.

Gorin et al[70]

SHED

In vivo

Mice

VEGF, FGF-2, HGF

Flow cytometry ELISA, and IHC

Cells

No

No

SHED has high angiogenic potential that hypoxia further increases

10.

Bento et al[71]

SHED

In vivo

Mice

VEGF

PCR

Cells

EGM-2MV supplemented with VEGF

No

Increased blood vessel formation

Periodontal ligament–derived mesenchymal stem cells (PDLSCs)

1.

Iwasaki et al[72]

PDL

In vitro

HUVECs

VEGF

ELISA

Conditioned media

No

HUVECs

HUVECs demonstrated minimal apoptotic activity on treatment with PDLSC-CM; increased vascular activity was noted at the same time

2.

Zhang et al[73]

PDL

In vitro

HUVECs

CD31 and VEGFA

Flow cytometry

No

No

HUVECs

HUVECs treated with exosomes derived from inflamed PDLSCs exhibited better tube formation than the control group

3.

Diomede et al[74]

PDL

In vitro

HUVECs

VEGF and RUNX2

Immunofluorescence and RT-PCR

Cells

Titanium surfaces, machined (CTRL) and dual acid-etched (TEST)

No

Human PDLSCs cultured on TEST evidenced a higher expression of VEGF and RUNX2 than hPDLSCs cultured on the CTRL surface

4.

Marconi et al[75]

PDL

In vitro

HUVECs

VEGF, VEGF-R, and RUNX2

Immunofluorescence

Cells

Titanium implant surfaces modified with two different procedures, sandblasted (control—CTRL) and sandblasted/etched (test—TEST), as experimental titanium surfaces

No

TEST surfaces compared to CTRL titanium surfaces enhanced cell adhesion and increased VEGF and RUNX2 expression

5.

Kim et al[76]

PDL

In vitro

HUVECs

VEGF, bFGF, and ANGPT1

PCR and Western blot analysis

Both

Cyclosporine A (CsA)

HUVECs

CsA reduced angiogenesis by blocking the ERK and p38/c-fos pathway in hPDLSCs

6.

Iwasaki et al[77]

PDL

In vivo

Rat

VEGF, bFGF, and HGF

Flow cytometry and PCR

Cells

No

No

VEGF expression was increased in PDLSCs

7.

Jearanaiphaisarn et al[78]

PDL

In vitro

HUVECs

VEGF, alpha-1 type I collagen (COL1), and essential bFGF

qPCR, ELISA, immunofluorescence staining

Cells

Iloprost, prostacyclin receptor (IP) antagonist

No

Iloprost promoted mRNA and protein expression of VEGF and COL1, but not of bFGF in hPDLSCs cells

8.

Wei et al[79]

PDL

In vitro

HUVECs

bFGF and Ang

PCR and flow cytometry

Cells

No

PDLSCs from healthy teeth and periodontally compromised teeth, rapamycin, and cDNA-Beclin-1

Proangiogenic cytokine expression increased, and more tube formation was observed in periodontally compromised teeth derived PDLSCs

9.

Bae et al[80]

PDL

In vivo

Mice

Stromal cell–derived factor 1 (SDF-1)

PCR and immunofluorescent

Cells

CXCR4 antagonist

HUVECs

Co-injection of PDLSCs and HUVECs worked up well for establishing vascular anastomosis

Stem cells from apical papilla (SCAPs)

1.

Yi et al[81]

SCAPs

In vivo

Mice

CD31, VEGFR2, VEGFR1, and TIE2

RT-PCR, western blotting, flow cytometry, and immunofluorescence

Cells

Acetylated low-density lipoprotein (ac-LDL)

HUVECs, SCAPs-endothelial cells

Angiogenic factors enhanced the differentiation of SCAPs into endothelial cells

2.

Liu et al[82]

SCAPs

In vitro

Endothelial cell lines

Hypoxia-inducible factor-1α (HIF-1α) and VEGF

RT-PCR and ELISA

Both

Cobalt-doped multiwalled carbon nanotube nanocomposites

Endothelial cells

Conditioned media collected from SCAP when treated with nanocomposites showed enhanced vessel formation

3.

Yu et al[83]

SCAPs

In vitro

HUVECs

VEGF and FGF-2

RT-PCR and immunofluorescence staining

Both

No

BM-MSCs, dental pulp cells (DPCs)

SCAPs-CM showed enhanced osteogenic and neurogenic differentiation in DPCs but did not prove to be significant in angiogenesis

4.

Yuan et al[84]

SCAPs

In vivo

Mice

VEGF

PCR and ELISA

Cells

SCAPs transduced with an ephrinB2-lentiviral expression vector (ephrinB2-SCAPs) in the experimental group and green fluorescent protein (GFP-SCAPs) in the control group

HUVECs

Enhanced expression of VEGF was observed with ephrinB2 transduction

5.

Koutsoumparis et al[85]

SCAPs

In vitro

HUVECs

PECAM-1, VEGFR2, vWF, and VE-cadherin/CDH5 MMP-2

RT-PCR and flow cytometric analysis

Cells

Recombinant human erythropoietin-alpha (rhEPOa)

No

rhEPOa is capable of promoting endothelial transdifferentiation of SCAP

6.

Yadlapati et al[86]

SCAPs

In vivo

Mice

Left-right determination factor 1 (LEFTY1), bone morphogenetic protein 8b (BMP8B), peptidylprolyl isomerase A (PPIA), bone morphogenetic protein 4 (BMP4), TGFβ1, FGF5, colony-stimulating factor 1 (CSF1), VEGFC, pleiotrophin (PTN), and ubiquitin C (UBC), VEGFA, PPIA, chemokine (C-X-C motif) ligand 1 (CXCL1), hydroxymethylbilane synthase (HMBS), RPL0, and inhibin beta A (INHBA)

ELISA

Cells

VEGF loaded (concentration of 12.2 ng/cm) polydioxanone fiber

No

Accelerated angiogenesis was achieved

7.

Yuan et al[87]

SCAPs

In vitro

HUVECs

VEGF

PCR and ELISA

Cells

EphrinB2

HUVECs

Co-culture of SCAPs and HUVECs accelerated the formation of vascularlike structures while inhibition of EphrinB2 expression suppressed the formation of vessel-like structures

8.

Peters et al[88]

SCAPs

In vitro

HUVECs

VEGF, ANGPT1, c-fos0-induced growth factor (FIGF), FGF2, and TGFβ1

Flow cytometry and PCR

Cells

ProRoot MTA or BD

No

VEGF expression was enhanced by stimulating either MTA or BD types of cement, but FGF and ANGPT1 expression were reduced

9.

Bakopoulou et al[89]

SCAPs

In vitro

HUVECs

Angiogenin, IGFBP-3, VEGF, PDGF, IGF1, MMPs, PECAM-1, and VE-cadherin

PCR, flow cytometry, and ELISA

Both

SCAP was exposed to serum deprivation (SD), glucose deprivation (GD), and oxygen deprivation/hypoxia (OD) conditions

HUVECs

Exposing the cells to stressed conditions proved to enhance the angiogenesis obtained from CM

10.

Yuan et al[90]

SCAPs

In vitro

HUVECs

VEGF, EphrinB2, angiopoietin, EphB4, insulin growth factor-1, EDN1, FGF, PDGF, and TGF-β

ELISA and RT-PCR

Cells

Hypoxic conditions

HUVECs

HIF-1a and ephrinB2 in SCAP under hypoxia are upregulated

Gingival mesenchymal stem cells (GMSCs)

1.

Jin et al[91]

GMSCs

In vivo

Mice

VEGF-A, TGF-β, and FGF-2

ELISA and RT-PCR

Both

Lentivirus transfection and FGF-2

HUVECs

FGF-2 gene-modified GMSCs constructed using lentiviral transfection promoted GMSCs paracrine of angiogenesis-related growth factors

Comparison of OC-MSC sources

1.

Zhu et al[92]

SHED and DPSC

In vitro

HUVECs

PDGFR-β, α-SMA, NG2, and DEMSIN

RT-PCR

Cells

No

HUVECs

DPSCs performed better as a candidate in angiogenic assays compared to SHED

2.

Xie et al[93]

SHED and DPMSC

In ovo

Chick embryo CAM

PECAM-1/CD31, VEGF, VEGF receptor 1 (VEGFR1), VEGF receptor 2 (VEGFR2), and vWF

RT-PCR

Cells

No

BM-MSCs

Angiogenic gene expressions were increased in SHED compared to DPMSCs or BM-MSCs

3.

Angelopoulos et al[94]

Gingival MSCs (GMSCs) and DPMSCs

In vivo

Mice

VEGF and HGF

Flow cytometry, ELISA, and IHC

Both

No

No

GMSCs showed an improved angiogenic capacity compared to DPMSCs

4.

Xu et al[95]

DPMSCs and SHED

In vivo

Mice

VEGF-A, VEGF-RI, PlGF-1, TGF-β, and SB-431542

RT-PCR and IHC

Cells

No

No

SHED possessed a higher endothelial differentiation potential than DPMSCs

5.

Osman et al[96]

PDLSCs and SHED

In vitro

HUVECs

TGF, IGF, FGF, VEGF, PDGF, and CTGF

PCR

Cells

No

No

PDLSCs showed a higher propensity toward angiogenesis compared to DPMSCs

Combined sources of stem cell

1.

Zhang et al[97]

DPSCs and SHED

In vivo

Mice

VEGFR2, Tie-2, CD31, and VE-cadherin

Flow cytometry

Cells

No

No

p53/p21 regulates the angiogenic potential of DPSCs and SHED in vivo

2.

Olcay et al[98]

DPMSCs, PDLSCs, and human tooth germ stem cells (hTGSCs)

In vitro

HUVECs

FGF-2, PDGF, and VEGF

Flow cytometry and ELISA

Both

Tricalcium silicate-based MRA (ProRoot MTA), BD, and a novel bioceramic root canal sealer (Well-Root ST) and Dycall are positive control groups

HUVECs

VEGF levels were significantly higher in a ProRoot MTA group

3.

Hilkens et al[99]

DPMSCs and SCAPs

In vivo

Mice

VEGF, primary bFGF, angiopoietin-1, MMPs, endostatin, thrombospondin-1, and IGFBP3

ELISA and IHC

Cells

No

No

Co-culture of DPMSCs and SCAPs provided enhanced angiogenic proliferation of cells and improved blood vessel growth in vivo

4.

Zhang et al[100]

DPMSCs and SHED

In vivo

Mice

VEGF, Wnt-β-catenin

PCR and IHC

Cells

No

No

Wnt/b-catenin silencing depressed angiogenesis by DPMSCs

5.

Hilkens et al[101]

SCAPs and DPMSCs

In ovo

Chorioallantoic membrane

VEGF, bFGF, HGF-1, ANGPT1, and IGFBP3

PCR and ELISA

Both

No

No

DPMSCs and SCAPs caused a significant increase in blood vessel count

The following data items were extracted: authors and year of publication; source of stem cells used; type of study; model used for evaluating angiogenesis; growth factors assessed for angiogenesis; method used for analysis of angiogenesis; use of stem cells/conditioned media; use of preconditioning; use of co-culture with MSCs; and results obtained.

To evaluate an article's quality, we used the Joanna Briggs Institute appraisal checklist for a case-control study. Based on 10 prespecified questions in the tool, two researchers independently examined all case reports. Each question received one of the following statuses based on judgment: “yes,” “no,” “maybe,” or “unclear.” A quality grade was assigned to the listed studies, with scores over 70% deemed excellent. Scores between 40 and 70% were considered to be of moderate quality, while those under 40% were considered to be of low quality. The reviewers agreed on these criteria in order to provide a thorough and objective assessment of the research quality. Egger's regression test was used to identify publication bias in the selected articles for quantitative analysis.


#

Results

In an initial literature search, 1,025 articles (591 from PubMed and 434 from Scopus) were retrieved. The selection strategy employed in the qualitative and quantitative analysis is illustrated using the PRISMA flowchart. The results of database searches were carefully maintained using Mendeley software (version 1803). Mendeley software (version 1803) for Windows (Elsevier, London, UK) was used in the initial phase of the screening process to remove duplicate articles. Five hundred and twenty-nine articles from both databases were excluded due to overlapping data. After scrutiny of the titles, 284 articles were selected. Abstracts and full texts of the remaining articles were further screened for relevance, and 80 articles were excluded. In addition, 70 reviews and letters to editors were excluded. Of the remaining 134 articles, 40 were excluded due to data being in languages other than English or irrelevance. Hence, a total of 94 articles were selected for data extraction. The data extracted from the included studies are summarized in [Table 1]. [Fig. 1] summarizes and depicts the PRISMA flowchart. Source-wise number of articles included in the review are depicted in the graph in [Fig. 2].

Zoom Image
Fig. 1 Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) chart illustrating the research methodology used in the review.
Zoom Image
Fig. 2 Graphical representation of the source-wise articles included in the review. DPSC, dental pulp stem cell; GMSC, gingival mesenchymal stem cell; PDLSC, periodontal ligament–derived mesenchymal stem cell; SCAP, stem cells from apical papilla; SHED, stem cells from human exfoliated deciduous teeth.

A quantitative analysis was possible only for comparing studies with an individual oral-derived source of MSCs. The DPMSCs and SCAPs were individually compared with the endothelial cell lines used to control in vitro analysis of blood vessel formation. Thus, the best source of oral-derived MSCs is not projected through the meta-analysis performed. Instead, the present meta-analysis shows that oral tissue–derived stem cells have more potential for augmenting angiogenesis than endothelial cell lines alone. The studies compared the in vitro tubule formation or total branching points between cases and controls. Out of the 94 studies in the systematic review, only four had data compatible with a meta-analysis.[34] [61] [84] [90] These studies referred to tubular formation's mean and total branch points in the case and control groups. The difference between the mean with standard deviation and the corresponding confidence interval was calculated for each study. Forest plots were created with RevMan software (version 5.4.1) using the calculated mean differences shown in [Figs. 3],[4],[5].

Zoom Image
Fig. 3 Summary of the meta-analysis assessing the effect of DPMSCs on the tubular length in an in vitro Matrigel assay showing a positive correlation of the co-culture of HUVEC and DPMSCs with the tubule length formation, which was statistically significant (p = 0.04). CI, confidence interval; DPMSCs, dental pulp–derived mesenchymal stem cells; DPSC, dental pulp stem cell; HUVEC, human umbilical vein endothelial cell; SD, standard deviation.
Zoom Image
Fig. 4 Summary of the meta-analysis assessing the effect of SCAP on the tubular length in an in vitro Matrigel assay showing a positive correlation of the co-culture of HUVEC and SCAPs with the tubule length formation, which was not statistically significant (p = 0.16). CI, confidence interval; HUVEC, human umbilical vein endothelial cell; SCAP, stem cells from apical papilla; SD, standard deviation.
Zoom Image
Fig. 5 Summary of the meta-analysis assessing the effect of SCAP on the total branching points in an in vitro Matrigel assay showing a positive correlation of the co-culture of HUVEC and SCAPs with the total branching point number, which was not statistically significant (p = 0.14). CI, confidence interval; HUVEC, human umbilical vein endothelial cell; SCAP, stem cells from apical papilla; SD, standard deviation.

The meta-analysis ([Fig. 3]) shows a positive correlation of the co-culture of human umbilical vein endothelial cells (HUVECs) and DPMSCs with tubule length formation, which was statistically significant (p = 0.04), with a mean difference of 0.20 and a 95% confidence interval of 0.01–0.40. Succeeding meta-analysis ([Figs. 4] and [5]) showed a positive correlation with the co-culture of HUVEC and the SCAP group with tubule length formation ([Fig. 4]) and total branching points ([Fig. 5]) with a mean difference of 5.20 and 20.78 and a 95% confidence interval of –2.05 to 12.45 and –6.66 to 48.21, respectively. Thus, the overall results from the meta-analysis revealed that oral-derived MSCs (DPSC and SCAP) carry a better angiogenic potential in vitro than the endothelial cell lines used alone, as depicted in the forest plot in [Figs. 3],[4],[5].

Assessment of Quality and Publication Bias

Ten of the 94 studies considered obtained ratings less than 70%, categorizing them as intermediate in quality. In contrast, the other 84 studies were classed as high quality given that their overall score surpassed 70%. The studies included for meta-analysis were high quality with score greater than 70%.

The Egger test showed a potential publication bias with 50% studies closer to the intercept line and 50% of studies away from the intercept line ([Table 2], [Fig. 6]). Such skewed results could be attributed to small sample of studies that were analyzed quantitatively.

Table 2

Tabular representation of Egger's regression test

Study reference

Z -score

SD

n

SE

1/SE

2012

2.27

0.7

3

0.404

2.474

2015

7.11

0.4

5

0.179

5.590

2016

30.23

0.45

3

0.260

3.849

2019

20.38

0.03

6

0.012

81.650

Abbreviations: SD, standard deviation; SE, standard error.


Zoom Image
Fig. 6 Graphical representation of Egger's regression test.

#
#

Discussion

After a detailed scrutiny of the literature, 94 articles meeting our inclusion criteria were included in the review, investigating the influence of MSCs or their secretomes derived from oral sources. Of these, 54 studies involved dental pulp, 10 articles investigated MSCs from SHED, and 9 investigated the PDL stem cells. The SCAPs were studied in 10 articles, and gingival MSCs (GMSCs) were explored in a single study. DPMSCs were relatively more explored for their angiogenic potential, as evidenced by the number of articles published. The critical parameters investigated to assess the effect of OC-MSCs and their secretomes on angiogenesis were tube capillary length and diameter, branching points, number of loops, expression of angiogenic proteins, endothelial cell proliferation in in vitro studies and capillary formation, enhanced wound healing, and generation of neovascularization in in ovo and in vivo studies. Postnatal MSCs (DPMSCs, PDL-derived stem cells [PDLSCs], SHED, GMSCs, and SCAP) retain the unique ability to form new functional blood vessels through angiogenesis.[97]

Dental Pulp–Derived Mesenchymal Stem Cells

The dental pulp is a rich source of MSCs that exhibit a self-renewal multilineage differentiation potential and secrete multiple proangiogenic factors. Thus, among the several therapeutic applications under investigation, the ability of DPMSCs to enhance angiogenesis has been the subject of active investigation.

Interestingly, the co-culture of DPMSCs with HUVECs exhibited a thick vessel-like structure, a characteristic feature of angiogenesis. The formation of vessel-like structures was absent in untreated HUVECs, confirming the angiogenic role of DPMSCs.[55] [61] DPMSCs could induce angiogenesis in a chicken chorioallantoic membrane model, as shown by the increased capillaries that observe a typical spoke wheel pattern around the DPMSCs Matrigel.[58] DPMSCs mediated noticeable repair of the infarcted myocardium in the animal model of myocardial infarction as an increase in the total number of blood vessels and an overall reduction in the infarct size was apparent. Therefore, the authors suggested DPMSCs as a potential alternative to bone marrow–derived MSCs to treat myocardial infarction.[59] [102] DPMSC-derived cells could promote neovasculogenesis in the mouse brain.[39]

Secretomes derived from DPMSCs have been actively investigated for their proangiogenic role. DPMSC secretomes also potentially enhance the proliferation of HUVECs.[24] DPMSC secretomes promoted angiogenesis in endothelial cell progenitors and terminally differentiated endothelial cells, as evidenced by the formation of tubelike structures in the Matrigel assay. In addition, DPMSC secretomes have been shown to improve the capillary density of skeletal muscles through improved angiogenesis, which can be attributed to the VEGF content in the secretomes. In the transwell migration assay performed on HUVECs, DPMSC secretomes promote better migration of HUVECs and microvascular network formation than the endothelial growth medium (EGM), suggesting a profound angiogenic role of DPMSC secretomes.[42] Under serum-free conditions, DPMSC secretomes have been shown to enhance the capillary tubelike formation from preexisting blood vessels, ultimately assisting angiogenesis.[29]

In a co-culture of secretomes derived from DPMSCs and bone marrow–derived MSCs, substantial proangiogenic changes were observed in the chorioallantoic membrane.[22] Furthermore, local intramuscular injection of DPMSC secretomes in the hindlimb ischemic mice model showed enhanced neovascularization and marked improved blood perfusion at the ischemic site.[54] [60] [62] Similar results were found in a mice model of ectopic tooth transplantation wherein enhanced expression of VEGF was noted, promoting pulp regeneration.[30] [77] Furthermore, DPMSC secretomes could promote pulplike vascularization in a scaffold implanted in a mouse model.[45]

One of the added therapeutic benefits of MSCs is their ability to secrete EV containing various nucleic acids, lipids, and proteins into the extracellular space. Many studies have suggested that EVs from MSCs can be employed for therapeutic applications in recent times. Interestingly, fibrin gel loaded with DPMSC-derived EVs enhanced cell migration and vascular tube formation in in vitro culture.[27] A mouse model was used to assess wound healing over the skin, where EVs derived from DPMSCs of healthy and periodontally compromised teeth were included. The results showed that EVs from DPMSCs from periodontally compromised teeth (P-DPMSCs) accelerated wound healing in mice compared to those derived from DPMSCs from healthy teeth.

Moreover, it showed enhanced blood vessel formation/angiogenesis, which forms the basis of wound healing, suggesting that the inflammatory microenvironment enhances the proangiogenic effects of DPMSCs. A comparative analysis between the DPMSCs derived from regular and deep carious teeth revealed that the expression levels of angiogenesis markers (VEGF, PDGF, stromal cell–derived growth factor-1) were higher in MSCs derived from deep carious pulp compared to the MSCs of the normal pulp. This suggests that an inflammatory microenvironment would instead work well for cell proliferation and further angiogenesis.[25] A combination of VEGF and IGF-1 enhances the angiogenic proliferation of DPMSCs from the carious environment synergistic effect.[32] Chronic inflammation-mediated tumor necrosis factor alpha induced initial apoptosis emerges DPSC into an angiogenic phenotype.[40] [56] The role of DPMSC EVs in angiogenesis is evident as miR-424 plays a regulatory role in angiogenesis.[57] Recently, modulation of the proangiogenic potential of DPMSCs by preconditioning, altering the culture conditions, and using novel biomaterials yielded promising results. Hypoxic preconditioning could enhance the proangiogenic capacity of DPMSCs.[43] [53] The expression of HIF-1α and SENP1 formed a positive feedback loop in angiogenesis promoted by DPMSCs under hypoxic conditions. HUVECs cultured with DPMSC secretomes treated with baicalein,[51] calcium phosphate cement (CPC), and CPC-bioactive glass nanoparticles (CPC-BGNs),[50] insulinlike growth factor binding protein 5 (IGFBP5)[10] exhibited higher expression of angiogenic markers in DPMSCs. DPMSCs treated with mineral trioxide aggregate (MTA), calcium hydroxide (Ca [OH]2), Biodentine (BD) and Emdogain,[23] EphrinB2-Fc, or EphB4-Fc[37] enhanced the expression of VEGF, which plays a crucial role in angiogenesis.[52]

In contrast, treatment with triethylene glycol dimethacrylate (TEGDMA) alone at a concentration of 0.25 mM downregulated the expression of angiogenic factors,[38] clindamycin and minocycline[35]; complete endothelial medium 2 (EGM-2) improved vessel formation; and angiogenic cell differentiation was achieved.[36] Aksel and Huang observed similar findings.[47] Treatment with 20% human platelet lysate under lipopolysaccharide-induced inflammatory environment in DPMSCs showed increased expression of proangiogenic markers.[6] Furthermore, the concentrated growth factor scaffold potentially enhanced endothelial cell proliferation and migration for DPMSCs.[41] Lipoprotein receptor–related protein signaling is required to express VEGF-promoting angiogenesis.[46] Decellularized matrix hydrogel derived from human dental pulp effectively promoted DPMSCs in a multidirectional differentiation.[31]


#

Stem Cells Obtained from Exfoliated Deciduous Teeth

SHED is a potent source of MSCs due to their higher proliferation potential, plasticity, and unique secretory profile. Few studies have explored the ability of SHED to enhance angiogenesis. Co-culture of the SHED with HUVECs promoted increased angiogenesis.[68] Furthermore, the SHED-HDMEC co-culture enhanced proangiogenic factor expression via NF-κB-dependent pathways.[65] Interestingly, SHED was subjected to shear stress-induced arterial endothelial differentiation.[4] SHED supplemented with an EGM showed augmented angiogenesis in vivo.[71] When subjected to a hypoxic environment, SHED augmented angiogenesis with improved function.[70] These studies suggest that SHED can be used as a perivascular source to form functional vascularlike structures in vivo.[76]


#

Periodontal Ligament–Derived Stem Cells

The PDL contains a population of progenitor cells, recently recognized as PDLSCs, capable of multilineage differentiation to produce tissues rich in collagen type I. Coadministration of PDLSCs and HUVECs showed anastomosis and enhanced blood vessel formation. It was seen that CXCR4 (an alpha-chemokine receptor specific for stromal-derived factor 1) antagonist inhibited blood vessel formation. This explains the role of PDLSCs in augmenting angiogenesis and blood vessel formation.[80] Furthermore, PDLSCs seeded on machined titanium disk surfaces showed increased VEGF expression, and RUNX2 (a gene inducing pluripotent stem cell differentiation to immature osteoblasts) plays a potential role in exhibiting angiogenesis.[74] In contrast, cyclosporine A–treated MSCs derived from PDL negatively impacted angiogenesis.[76]

Furthermore, prostacyclin pretreated PDL stem cells negatively impacted iloprost enhanced angiogenic marker expression.[78] PDLSCs derived from healthy and inflamed tissue (periodontally compromised teeth) were subjected to proliferation and angiogenesis. The results depicted that the inflammatory microenvironment provided better augmentation for angiogenesis, which agrees with the findings on DPMSCs.[73] [79]


#

Stem Cells Derived from Apical Papilla

A unique population of SCAP of the growing tooth root tips with embryoniclike properties is readily accessible in dental clinical practice from extracted wisdom teeth. Exposure of SCAP to various stress microenvironments and their respective secretomes has promoted angiogenesis.[89] EphrinB2 (a transmembrane ligand of EphB receptor tyrosine kinases expressed explicitly in arteries) could stabilize the vessel-like structure generated by the co-culture of SCAPs and HUVECs in vitro.[87] Co-culture of HUVECs and SCAPs under hypoxic conditions promoted the formation of endothelial tubules and a blood capillary network, which was in agreement with those obtained by Nam et al.[49] VEGF-loaded fibers can be considered a viable option for stimulating SCAP angiogenesis and new histogenesis during the endodontic procedure.[86] EphrinB2-transduced SCAPs could express VEGF marker in numerous amounts compared to the control group; its co-culture with HUVECs showed enhanced blood vessel formation in a Matrigel plug assay.[84] Treatment of SCAP cells with recombinant human erythropoietin-alpha (rhEPOa) elicits a proangiogenesis program by activating the Erythropoetin Receptor pathway.[85] Exposure of SCAP to MTA and BD (root-end filling material used in endodontic therapy of root canals) stimulated angiogenic gene expression and VEGF release inducing similar expression patterns in both MTA and BD. However, they appear to inhibit the expression of specific genes, including ANGPT1 and FGF2.[88] SCAP-derived secretomes improved osteogenic and neurogenic differentiation of dental pulp cells, but angiogenic differentiation did not significantly improve.[83]


#

Stem Cells Derived from Gingiva

The gingiva of human dentition is blessed with a remarkable contribution of neural crest ectomesenchyme, perifollicular mesenchyme, and partly the dental follicle proper. The origin of this tissue and its close approximation with the tooth give the GMSCs an exclusive position to stand apart from the rest of the oral cavity–derived cells. A study by Jin et al showed that when GMSCs were transfected with FGF-2, their expression potential for VEGF and TGF-β increased. Also, the secretomes derived from untreated GMSCs enhanced the gene and protein expression of angiogenic-related factors, endothelial tube formation, and cell migration capacity. However, the results obtained had an inferior efficacy than those obtained by the transfected GMSCs and their secretomes.[91]

Several researchers have investigated the comparative potential of OC-MSCs to explore the ideal source of MSCs in the augmentation of angiogenesis. In a study by Angelopoulos et al, GMSCs potentially proliferate, migrate, and form angiogenic tubules better than DPMSCs in vitro and in vivo.[94] Another study performed by Xu et al compared SHED and DPMSCs in enhancing angiogenesis. Their findings revealed that SHED possesses better angiogenic potential than the DPMSCs.[95] Furthermore, SHED showed a more substantial angiogenesis differentiation and proliferation potential than DPMSCs. Furthermore, PDLSCs exhibited better angiogenic potential than DPMSCs.[96] However, very few studies have reported the comparative potential of OC-MSCs.

In yet another study, a co-culture of DPMSCs and SCAPs exhibited improved blood vessel formation in vivo.[99] Furthermore, in an in ovo angiogenesis assay, the co-culture of DPMSCs and SCAPs showed better angiogenesis than the single source.[101] A root canal obturating material, Well-Root ST stimulated neovascularization during endodontic regeneration procedures. Furthermore, Well-Root ST showed better efficacy than BD or ProRoot MTA for stimulation in various oral-derived MSCs (DPMSCs, SHED, PDLSCs, GMSCs, and SCAP).[98]

The field of oral cavity–derived stem cells, particularly MSCs from dental pulp and apical papilla, has garnered interest due to their unique characteristics and potential applications in regenerative medicine. The finding that these stem cells have strong angiogenic potential holds several clinical implications and suggests promising directions for future research that could benefit the population in various ways.

  • Tissue regeneration: The angiogenic potential of oral cavity–derived stem cells suggests their capability to stimulate the formation of new blood vessels. This can be extremely valuable in regenerating damaged tissues, such as those affected by injury, disease, or degeneration. These stem cells could aid in promoting blood supply and nutrients to the regenerating tissue, enhancing the overall healing process.

  • Wound healing: The ability of these stem cells to promote angiogenesis can significantly accelerate wound healing in various clinical scenarios. For instance, they could be employed in chronic wound management, diabetic ulcer treatment, and postsurgical wound healing to expedite tissue repair and reduce complications.

  • Bone regeneration: Oral-derived MSCs have shown potential for bone tissue regeneration. Enhancing angiogenesis could aid in developing more effective treatments for bone defects, fractures, and conditions like osteoporosis.

  • Dental applications: The dental pulp and apical papilla are easily accessible sources of MSCs. This accessibility could make these stem cells valuable for various dental applications, such as periodontal tissue regeneration, dental implant support, and treatment of oral diseases.

  • Cardiovascular disorders: Given their angiogenic properties, these stem cells might hold promise in treating cardiovascular diseases. They could stimulate the growth of new blood vessels in ischemic heart tissue, potentially reducing the impact of heart attacks.


#
#

Limitations

The current literature shows a paucity of studies involving sources other than dental pulp. Even though OC-MSCs have proved their enhanced potential compared to other MSCs, further target-oriented comprehensive research is required to conclude which oral-derived stem cells have the most significant angiogenic potential. The systematic review involves different oral sources for MSCs, where maximum studies include dental pulp, and data for other sources (SHED, PDLSC, SCAP, and GMSC) are limited; therefore, a comparative evaluation could not be done. This systematic review incorporates in vitro, ex vivo, and in vivo trials and the data appear to be skewed. One specific type of research design might be advocated for better outcomes.


#

Conclusion

The specific objectives of our study were to explore whether easily accessible OC-MSCs from dental pulp and apical papilla had good angiogenic potential. The reviewed literature shows that all the OC-MSCs augmented angiogenesis in various experiments. In the studies comparing DPMSCs and PDLSC, GMSCs, or SHED, the latter sources have shown increased significant potential for angiogenesis compared to that of the DPMSCs. MSCs obtained from different places show close phenotypic characteristics. However, it is still unclear how similar they are since proliferation and differentiation capabilities in the presence of different growth factor stimuli differ depending on the source of origin. For instance, bone marrow MSCs tend to lose their proliferative potential with age. DPSCs, on the other hand, have a higher proliferation index and growth potential. DPSCs show the highest odontogenic capability under the same inductive microenvironment in comparison to bone marrow stromal stem cells.[103]

Avenues that can be explored further in the research realm are angiogenesis mechanisms, optimal delivery methods, combination therapy, and personalized medicine. This knowledge of precise molecular and cellular mechanisms underlying the angiogenic potential of oral-derived MSCs could lead to the development of targeted therapies. Future research could focus on identifying the most effective methods for delivering oral-derived MSCs to target tissues. This could involve investigating various delivery vehicles, such as scaffolds or hydrogels, to ensure the stem cells reach their intended destination. Furthermore, research might delve into tailoring treatments based on individual patient characteristics to maximize the regenerative potential. Exploring combination therapies, such as coupling oral-derived MSCs with growth factors or other regenerative agents, could enhance their angiogenic potential and effectiveness in various applications. Regenerative medicine and stem cells will usher in a renaissance in therapy in the near future.

The manuscript has been checked with the Fi-index tool and obtained a scrore of 0.60 for the first author on September 3, 2023 according to the Scopus database. The Fi-index tool aims to ensure the quality of the reference list and limit autocitations.[104] [105]


#
#

Conflict of Interest

None declared.

Authors' Contribution

All the authors contributed to the concept and design of the study.


Data Availability Statement

The datasets generated and/or analyzed during the current study are available from the corresponding author on request.


  • References

  • 1 Honnegowda TM, Kumar P, Udupa EG, Kumar S, Kumar U, Rao P. Role of angiogenesis and angiogenic factors in acute and chronic wound healing. Plast Aesthet Res 2015; 2: 243-249
  • 2 Auerbach R, Auerbach W, Polakowski I. Assays for angiogenesis: a review. Pharmacol Ther 1991; 51 (01) 1-11
  • 3 Adair TH, Montani JP. Angiogenesis. In: Granger DN, Granger JP. eds. Colloquium Series on Integrated Systems Physiology: From Molecule to Function. San Rafael, CA: Morgan & Claypool Life Sciences; 2010: 1-84
  • 4 Wang P, Zhu S, Yuan C, Wang L, Xu J, Liu Z. Shear stress promotes differentiation of stem cells from human exfoliated deciduous teeth into endothelial cells via the downstream pathway of VEGF-Notch signaling. Int J Mol Med 2018; 42 (04) 1827-1836
  • 5 King A, Balaji S, Keswani SG, Crombleholme TM. The role of stem cells in wound angiogenesis. Adv Wound Care (New Rochelle) 2014; 3 (10) 614-625
  • 6 Bindal P, Gnanasegaran N, Bindal U. et al. Angiogenic effect of platelet-rich concentrates on dental pulp stem cells in inflamed microenvironment. Clin Oral Investig 2019; 23 (10) 3821-3831
  • 7 Chakraborty S, Ponrasu T, Chandel S, Dixit M, Muthuvijayan V. Reduced graphene oxide-loaded nanocomposite scaffolds for enhancing angiogenesis in tissue engineering applications. R Soc Open Sci 2018; 5 (05) 172017
  • 8 Kerkis I, Kerkis A, Dozortsev D. et al. Isolation and characterization of a population of immature dental pulp stem cells expressing OCT-4 and other embryonic stem cell markers. Cells Tissues Organs 2006; 184 (3–4): 105-116
  • 9 Miran S, Mitsiadis TA, Pagella P. Innovative dental stem cell-based research approaches: the future of dentistry. Stem Cells Int 2016; 2016: 7231038
  • 10 Li J, Zhu Y, Li N. et al. Upregulation of ETV2 expression promotes endothelial differentiation of human dental pulp stem cells. Cell Transplant 2021; 30: 963689720978739
  • 11 Boreak N, Khayrat NMA, Shami AO. et al. Metformin pre-conditioning enhances the angiogenic ability of the secretome of dental pulp stem cells. Saudi Pharm J 2021; 29 (08) 908-913
  • 12 Li Y, Zhang Y, Wang H. et al. Dental pulp mesenchymal stem cells attenuate limb ischemia via promoting capillary proliferation and collateral development in a preclinical model. Stem Cells Int 2021; 2021: 5585255
  • 13 Li M, Wang Q, Han Q. et al. Novel molecule Nell-1 promotes the angiogenic differentiation of dental pulp stem cells. Front Physiol 2021; 12: 703593
  • 14 Alghutaimel H, Yang X, Drummond B, Nazzal H, Duggal M, Raïf E. Investigating the vascularization capacity of a decellularized dental pulp matrix seeded with human dental pulp stem cells: in vitro and preliminary in vivo evaluations. Int Endod J 2021; 54 (08) 1300-1316
  • 15 Zhou H, Li X, Wu RX. et al. Periodontitis-compromised dental pulp stem cells secrete extracellular vesicles carrying miRNA-378a promote local angiogenesis by targeting Sufu to activate the Hedgehog/Gli1 signalling. Cell Prolif 2021; 54 (05) e13026
  • 16 Huang X, Qiu W, Pan Y. et al. Exosomes from LPS-stimulated hDPSCs activated the angiogenic potential of HUVECs in vitro . Stem Cells Int 2021; 2021: 6685307
  • 17 Afami ME, El Karim I, About I, Coulter SM, Laverty G, Lundy FT. Ultrashort peptide hydrogels display antimicrobial activity and enhance angiogenic growth factor release by dental pulp stem/stromal cells. Materials (Basel) 2021; 14 (09) 2237
  • 18 Liao ZH, Zhu HQ, Chen YY. et al. The epigallocatechin gallate derivative Y6 inhibits human hepatocellular carcinoma by inhibiting angiogenesis in MAPK/ERK1/2 and PI3K/AKT/ HIF-1α/VEGF dependent pathways. J Ethnopharmacol 2020; 259: 112852
  • 19 He Y, Cao Y, Xiang Y. et al. An evaluation of norspermidine on anti-fungal effect on mature Candida albicans biofilms and angiogenesis potential of dental pulp stem cells. Front Bioeng Biotechnol 2020; 8: 948
  • 20 Guo S, Redenski I, Landau S, Szklanny A, Merdler U, Levenberg S. Prevascularized scaffolds bearing human dental pulp stem cells for treating complete spinal cord injury. Adv Healthc Mater 2020; 9 (20) e2000974
  • 21 Luzuriaga J, Irurzun J, Irastorza I, Unda F, Ibarretxe G, Pineda JR. Vasculogenesis from human dental pulp stem cells grown in matrigel with fully defined serum-free culture media. Biomedicines 2020; 8 (11) 483
  • 22 Merckx G, Hosseinkhani B, Kuypers S. et al. Angiogenic effects of human dental pulp and bone marrow-derived mesenchymal stromal cells and their extracellular vesicles. Cells 2020; 9 (02) 312
  • 23 Caseiro AR, Santos Pedrosa S, Ivanova G. et al. Mesenchymal Stem/ Stromal Cells metabolomic and bioactive factors profiles: a comparative analysis on the umbilical cord and dental pulp derived stem/stromal cells secretome. PLoS One 2019; 14 (11) e0221378
  • 24 Makino E, Nakamura N, Miyabe M. et al. Conditioned media from dental pulp stem cells improved diabetic polyneuropathy through anti-inflammatory, neuroprotective and angiogenic actions: cell-free regenerative medicine for diabetic polyneuropathy. J Diabetes Investig 2019; 10 (05) 1199-1208
  • 25 Chen Y, Li X, Wu J, Lu W, Xu W, Wu B. Dental pulp stem cells from human teeth with deep caries displayed an enhanced angiogenesis potential in vitro . J Dent Sci 2021; 16 (01) 318-326
  • 26 Li J, Rao Z, Zhao Y. et al. A decellularized matrix hydrogel derived from human dental pulp promotes dental pulp stem cell proliferation, migration, and induced multidirectional differentiation in vitro . J Endod 2020; 46 (10) 1438-1447.e5
  • 27 Wang D, Lyu Y, Yang Y. et al. Schwann cell-derived EVs facilitate dental pulp regeneration through endogenous stem cell recruitment via SDF-1/CXCR4 axis. Acta Biomater 2022; 140: 610-624
  • 28 Zhou J, Sun C. SENP1/HIF-1α axis works in angiogenesis of human dental pulp stem cells. J Biochem Mol Toxicol 2020; 34 (03) e22436
  • 29 Qu C, Brohlin M, Kingham PJ, Kelk P. Evaluation of growth, stemness, and angiogenic properties of dental pulp stem cells cultured in cGMP xeno-/serum-free medium. Cell Tissue Res 2020; 380 (01) 93-105
  • 30 Zhu L, Dissanayaka WL, Zhang C. Dental pulp stem cells overexpressing stromal-derived factor-1α and vascular endothelial growth factor in dental pulp regeneration. Clin Oral Investig 2019; 23 (05) 2497-2509
  • 31 Li J, Diao S, Yang H, Cao Y, Du J, Yang D. IGFBP5 promotes angiogenic and neurogenic differentiation potential of dental pulp stem cells. Dev Growth Differ 2019; 61 (09) 457-465
  • 32 Lu W, Xu W, Li J, Chen Y, Pan Y, Wu B. Effects of vascular endothelial growth factor and insulin growth factor–1 on proliferation, migration, osteogenesis and vascularization of human carious dental pulp stem cells. Mol Med Rep 2019; 20 (04) 3924-3932
  • 33 Youssef AR, Emara R, Taher MM. et al. Effects of mineral trioxide aggregate, calcium hydroxide, Biodentine and Emdogain on osteogenesis, Odontogenesis, angiogenesis and cell viability of dental pulp stem cells. BMC Oral Health 2019; 19 (01) 133
  • 34 Rapino M, Di Valerio V, Zara S. et al. Chitlac-coated thermosets enhance osteogenesis and angiogenesis in a co-culture of dental pulp stem cells and endothelial cells. Nanomaterials (Basel) 2019; 9 (07) 928
  • 35 Dubey N, Xu J, Zhang Z, Nör JE, Bottino MC. Comparative evaluation of the cytotoxic and angiogenic effects of minocycline and clindamycin: an in vitro study. J Endod 2019; 45 (07) 882-889
  • 36 Delle Monache S, Martellucci S, Clementi L. et al. In vitro conditioning determines the capacity of dental pulp stem cells to function as pericyte-like cells. Stem Cells Dev 2019; 28 (10) 695-706
  • 37 Gong T, Xu J, Heng B. et al. EphrinB2/EphB4 signaling regulates DPSCs to induce sprouting angiogenesis of endothelial cells. J Dent Res 2019; 98 (07) 803-812
  • 38 Schertl P, Volk J, Perduns R. et al. Impaired angiogenic differentiation of dental pulp stem cells during exposure to the resinous monomer triethylene glycol dimethacrylate. Dent Mater 2019; 35 (01) 144-155
  • 39 Luzuriaga J, Pastor-Alonso O, Encinas JM, Unda F, Ibarretxe G, Pineda JR. Human dental pulp stem cells grown in neurogenic media differentiate into endothelial cells and promote neovasculogenesis in the mouse brain. Front Physiol 2019; 10: 347
  • 40 Zou T, Jiang S, Dissanayaka WL. et al. Sema4D/PlexinB1 promotes endothelial differentiation of dental pulp stem cells via activation of AKT and ERK1/2 signaling. J Cell Biochem 2019; 120 (08) 13614-13624
  • 41 Jin R, Song G, Chai J, Gou X, Yuan G, Chen Z. Effects of concentrated growth factor on proliferation, migration, and differentiation of human dental pulp stem cells in vitro . J Tissue Eng 2018; 9: 2041731418817505
  • 42 Gharaei MA, Xue Y, Mustafa K, Lie SA, Fristad I. Human dental pulp stromal cell conditioned medium alters endothelial cell behavior. Stem Cell Res Ther 2018; 9 (01) 69
  • 43 Dou L, Yan Q, Liang P, Zhou P, Zhang Y, Ji P. iTRAQ-based proteomic analysis exploring the influence of hypoxia on the proteome of dental pulp stem cells under 3D culture. Proteomics 2018; 18 (3–4): 1700215
  • 44 Aksel H, Öztürk Ş, Serper A, Ulubayram K. VEGF/BMP-2 loaded three-dimensional model for enhanced angiogenic and odontogenic potential of dental pulp stem cells. Int Endod J 2018; 51 (04) 420-430
  • 45 Lambrichts I, Driesen RB, Dillen Y. et al. Dental pulp stem cells: their potential in reinnervation and angiogenesis by using scaffolds. J Endod 2017; 43 (9S): S12-S16
  • 46 Silva GO, Zhang Z, Cucco C, Oh M, Camargo CHR, Nör JE. Lipoprotein receptor–related protein 6 signaling is necessary for vasculogenic differentiation of human dental pulp stem cells. J Endod 2017; 43 (9S): S25-S30
  • 47 Aksel H, Huang GT. Human and swine dental pulp stem cells form a vascularlike network after angiogenic differentiation in comparison with endothelial cells: a quantitative analysis. J Endod 2017; 43 (04) 588-595
  • 48 Zou T, Dissanayaka WL, Jiang S. et al. Semaphorin 4D enhances angiogenic potential and suppresses osteo-/odontogenic differentiation of human dental pulp stem cells. J Endod 2017; 43 (02) 297-305
  • 49 Nam H, Kim GH, Bae YK. et al. Angiogenic capacity of dental pulp stem cell regulated by SDF-1 α-CXCR4 axis. Stem Cells Int 2017; 2017: 8085462
  • 50 Lee SI, Lee ES, El-Fiqi A, Lee SY, Eun-Cheol Kim Kim HW. Stimulation of odontogenesis and angiogenesis via bioactive nanocomposite calcium phosphate cements through integrin and VEGF signaling pathways. J Biomed Nanotechnol 2016; 12 (05) 1048-1062
  • 51 Lee SI, Kim SY, Park KR, Kim EC. Baicalein promotes angiogenesis and odontoblastic differentiation via the BMP and Wnt pathways in human dental pulp cells. Am J Chin Med 2016; 44 (07) 1457-1472
  • 52 Spina A, Montella R, Liccardo D. et al. NZ-GMP approved serum improve hDPSC osteogenic commitment and increase angiogenic factor expression. Front Physiol 2016; 7: 354
  • 53 Kuang R, Zhang Z, Jin X. et al. Nanofibrous spongy microspheres for the delivery of hypoxia-primed human dental pulp stem cells to regenerate vascularized dental pulp. Acta Biomater 2016; 33: 225-234
  • 54 Shen C, Li L, Feng T. et al. Dental pulp stem cells derived conditioned medium promotes angiogenesis in hindlimb ischemia. J Tissue Eng Regen Med 2015; 12 (01) 59-68
  • 55 Dissanayaka WL, Zhu L, Hargreaves KM, Jin L, Zhang C. In vitro analysis of scaffold-free prevascularized microtissue spheroids containing human dental pulp cells and endothelial cells. J Endod 2015; 41 (05) 663-670
  • 56 Boyle M, Chun C, Strojny C. et al. Chronic inflammation and angiogenic signaling axis impairs differentiation of dental-pulp stem cells. PLoS One 2014; 9 (11) e113419
  • 57 Liu W, Gong Q, Ling J, Zhang W, Liu Z, Quan J. Role of miR-424 on angiogenic potential in human dental pulp cells. J Endod 2014; 40 (01) 76-82
  • 58 Bronckaers A, Hilkens P, Fanton Y. et al. Angiogenic properties of human dental pulp stem cells. PLoS One 2013; 8 (08) e71104
  • 59 Janebodin K, Zeng Y, Buranaphatthana W, Ieronimakis N, Reyes M. VEGFR2-dependent angiogenic capacity of pericyte-like dental pulp stem cells. J Dent Res 2013; 92 (06) 524-531
  • 60 Ishizaka R, Hayashi Y, Iohara K. et al. Stimulation of angiogenesis, neurogenesis and regeneration by side population cells from dental pulp. Biomaterials 2013; 34 (08) 1888-1897
  • 61 Dissanayaka WL, Zhan X, Zhang C, Hargreaves KM, Jin L, Tong EH. Coculture of dental pulp stem cells with endothelial cells enhances osteo-/odontogenic and angiogenic potential in vitro . J Endod 2012; 38 (04) 454-463
  • 62 Iohara K, Zheng L, Wake H. et al. A novel stem cell source for vasculogenesis in ischemia: subfraction of side population cells from dental pulp. Stem Cells 2008; 26 (09) 2408-2418
  • 63 Wu M, Liu X, Li Z. et al. SHED aggregate exosomes shuttled miR-26a promote angiogenesis in pulp regeneration via TGF-β/SMAD2/3 signalling. Cell Prolif 2021; 54 (07) e13074
  • 64 Han Y, Chen Q, Zhang L, Dissanayaka WL. Indispensable role of HIF-1α signaling in post-implantation survival and angio-/vasculogenic properties of SHED. Front Cell Dev Biol 2021; 9: 655073
  • 65 Zaw SYM, Kaneko T, Zaw ZCT. et al. Crosstalk between dental pulp stem cells and endothelial cells augments angiogenic factor expression. Oral Dis 2020; 26 (06) 1275-1283
  • 66 Atlas Y, Gorin C, Novais A. et al. Microvascular maturation by mesenchymal stem cells in vitro improves blood perfusion in implanted tissue constructs. Biomaterials 2021; 268: 120594
  • 67 Guo H, Zhao W, Liu A. et al. SHED promote angiogenesis in stem cell-mediated dental pulp regeneration. Biochem Biophys Res Commun 2020; 529 (04) 1158-1164
  • 68 Gong T, Heng BC, Xu J. et al. Decellularized extracellular matrix of human umbilical vein endothelial cells promotes endothelial differentiation of stem cells from exfoliated deciduous teeth. J Biomed Mater Res A 2017; 105 (04) 1083-1093
  • 69 Kim JH, Kim GH, Kim JW. et al. In vivo angiogenic capacity of stem cells from human exfoliated deciduous teeth with human umbilical vein endothelial cells. Mol Cells 2016; 39 (11) 790-796
  • 70 Gorin C, Rochefort GY, Bascetin R. et al. Priming dental pulp stem cells with fibroblast growth factor-2 increases angiogenesis of implanted tissue-engineered constructs through hepatocyte growth factor and vascular endothelial growth factor secretion. Stem Cells Transl Med 2016; 5 (03) 392-404
  • 71 Bento LW, Zhang Z, Imai A. et al. Endothelial differentiation of SHED requires MEK1/ERK signaling. J Dent Res 2013; 92 (01) 51-57
  • 72 Iwasaki K, Akazawa K, Nagata M. et al. Angiogenic effects of secreted factors from periodontal ligament stem cells. Dent J 2021; 9 (01) 9
  • 73 Zhang Z, Shuai Y, Zhou F. et al. PDLSCs regulate angiogenesis of periodontal ligaments via VEGF transferred by exosomes in periodontitis. Int J Med Sci 2020; 17 (05) 558-567
  • 74 Diomede F, Marconi GD, Cavalcanti MFXB. et al. VEGF/VEGF-R/RUNX2 upregulation in human periodontal ligament stem cells seeded on dual acid etched titanium disk. Materials (Basel) 2020; 13 (03) 706
  • 75 Marconi GD, Diomede F, Pizzicannella J. et al. Enhanced VEGF/VEGF-R and RUNX2 expression in human periodontal ligament stem cells cultured on sandblasted/etched titanium disk. Front Cell Dev Biol 2020; 8: 315
  • 76 Kim HJ, Yoo JH, Choi Y, Joo JY, Lee JY, Kim HJ. Assessing the effects of cyclosporine A on the osteoblastogenesis, osteoclastogenesis, and angiogenesis mediated by human periodontal ligament stem cells. J Periodontol 2020; 91 (06) 836-848
  • 77 Iwasaki K, Nagata M, Akazawa K, Watabe T, Morita I. Changes in characteristics of periodontal ligament stem cells in spheroid culture. J Periodontal Res 2019; 54 (04) 364-373
  • 78 Jearanaiphaisarn T, Sanharati T, Pavasant P, Nakalekha Limjeerajarus C. The effect of iloprost on cell proliferation and angiogenesis-related gene expression in human periodontal ligament cells. Odontology 2018; 106 (01) 11-18
  • 79 Wei W, An Y, An Y, Fei D, Wang Q. Activation of autophagy in periodontal ligament mesenchymal stem cells promotes angiogenesis in periodontitis. J Periodontol 2018; 89 (06) 718-727
  • 80 Bae YK, Kim GH, Lee JC. et al. The significance of SDF-1α-CXCR4 axis in in vivo angiogenic ability of human periodontal ligament stem cells. Mol Cells 2017; 40 (06) 386-392
  • 81 Yi B, Ding T, Jiang S. et al. Conversion of stem cells from apical papilla into endothelial cells by small molecules and growth factors. Stem Cell Res Ther 2021; 12 (01) 266
  • 82 Liu J, Zou T, Yao Q, Zhang Y, Zhao Y, Zhang C. Hypoxia-mimicking cobalt-doped multi-walled carbon nanotube nanocomposites enhance the angiogenic capacity of stem cells from apical papilla. Mater Sci Eng C 2021; 120: 111797
  • 83 Yu S, Zhao Y, Fang TJ, Ge L. Effect of the soluble factors released by dental apical papilla-derived stem cells on the osteo/odontogenic, angiogenic, and neurogenic differentiation of dental pulp cells. Stem Cells Dev 2020; 29 (12) 795-805
  • 84 Yuan C, Wang P, Zhu S. et al. Overexpression of ephrinB2 in stem cells from apical papilla accelerates angiogenesis. Oral Dis 2019; 25 (03) 848-859
  • 85 Koutsoumparis A, Vassili A, Bakopoulou A, Ziouta A, Tsiftsoglou AS. Erythropoietin (rhEPOa) promotes endothelial transdifferentiation of stem cells of the apical papilla (SCAP). Arch Oral Biol 2018; 96: 96-103
  • 86 Yadlapati M, Biguetti C, Cavalla F. et al. Characterization of a vascular endothelial growth factor–loaded bioresorbable delivery system for pulp regeneration. J Endod 2017; 43 (01) 77-83
  • 87 Yuan C, Wang P, Zhu S. et al. EphrinB2 stabilizes vascularlike structures generated by endothelial cells and stem cells from apical papilla. J Endod 2016; 42 (09) 1362-1370
  • 88 Peters OA, Galicia J, Arias A, Tolar M, Ng E, Shin SJ. Effects of two calcium silicate cements on cell viability, angiogenic growth factor release and related gene expression in stem cells from the apical papilla. Int Endod J 2016; 49 (12) 1132-1140
  • 89 Bakopoulou A, Kritis A, Andreadis D. et al. Angiogenic potential and secretome of human apical papilla mesenchymal stem cells in various stress microenvironments. Stem Cells Dev 2015; 24 (21) 2496-2512
  • 90 Yuan C, Wang P, Zhu L. et al. Coculture of stem cells from apical papilla and human umbilical vein endothelial cell under hypoxia increases the formation of three-dimensional vessel-like structures in vitro . Tissue Eng Part A 2015; 21 (5–6): 1163-1172
  • 91 Jin S, Yang C, Huang J. et al. Conditioned medium derived from FGF-2-modified GMSCs enhances migration and angiogenesis of human umbilical vein endothelial cells. Stem Cell Res Ther 2020; 11 (01) 68
  • 92 Zhu SY, Yuan CY, Lin YF. et al. Stem cells from human exfoliated deciduous teeth (SHEDs) and dental pulp stem cells (DPSCs) display a similar profile with pericytes. Stem Cells Int 2021; 2021: 8859902
  • 93 Xie J, Zhao YM, Rao NQ. et al. Comparative study of differentiation potential of mesenchymal stem cells derived from orofacial system into vascular endothelial cells. Beijing Da Xue Xue Bao Yi Xue Ban 2019; 51 (05) 900-906
  • 94 Angelopoulos I, Brizuela C, Khoury M. Gingival mesenchymal stem cells outperform haploidentical dental pulp-derived mesenchymal stem cells in proliferation rate, migration ability, and angiogenic potential. Cell Transplant 2018; 27 (06) 967-978
  • 95 Xu JG, Gong T, Wang YY. et al. Inhibition of TGF-β signaling in SHED enhances endothelial differentiation. J Dent Res 2018; 97 (02) 218-225
  • 96 Osman A, Gnanasegaran N, Govindasamy V. et al. Basal expression of growth-factor-associated genes in periodontal ligament stem cells reveals multiple distinctive pathways. Int Endod J 2014; 47 (07) 639-651
  • 97 Zhang Z, Oh M, Sasaki JI, Nör JE. Inverse and reciprocal regulation of p53/p21 and Bmi-1 modulates vasculogenic differentiation of dental pulp stem cells. Cell Death Dis 2021; 12 (07) 644
  • 98 Olcay K, Taşli PN, Güven EP. et al. Effect of a novel bioceramic root canal sealer on the angiogenesis-enhancing potential of assorted human odontogenic stem cells compared with principal tricalcium silicate-based cements. J Appl Oral Sci 2020; 28: e20190215
  • 99 Hilkens P, Bronckaers A, Ratajczak J, Gervois P, Wolfs E, Lambrichts I. The angiogenic potential of DPSCs and SCAPs in an in vivo model of dental pulp regeneration. Stem Cells Int 2017; 2017: 2582080
  • 100 Zhang Z, Nör F, Oh M, Cucco C, Shi S, Nör JE. Wnt/β-catenin signaling determines the vasculogenic fate of postnatal mesenchymal stem cells. Stem Cells 2016; 34 (06) 1576-1587
  • 101 Hilkens P, Fanton Y, Martens W. et al. Pro-angiogenic impact of dental stem cells in vitro and in vivo . Stem Cell Res (Amst) 2014; 12 (03) 778-790
  • 102 Gandia C, Armiñan A, García-Verdugo JM. et al. Human dental pulp stem cells improve left ventricular function, induce angiogenesis, and reduce infarct size in rats with acute myocardial infarction. Stem Cells 2008; 26 (03) 638-645
  • 103 Zeidán-Chuliá F, Noda M. “Opening” the mesenchymal stem cell tool box. Eur J Dent 2009; 3 (03) 240-249
  • 104 Fiorillo L. Fi-index: a new method to evaluate authors Hirsch-index reliability. Publ Res Q 2022; 38: 465-474
  • 105 Fiorillo L, Cicciù M. The use of Fi-index tool to assess per-manuscript self-citations. Publ Res Q 2022; 38: 684-692

Address for correspondence

Yash Merchant, MDS, FRCS
Consultant Head and Neck Surgeon, Department of Oral and Maxillofacial Surgery, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth
Pimpri, Pune
India   

Publication History

Article published online:
23 November 2023

© 2023. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution License, permitting unrestricted use, distribution, and reproduction so long as the original work is properly cited. (https://creativecommons.org/licenses/by/4.0/)

Thieme Medical and Scientific Publishers Pvt. Ltd.
A-12, 2nd Floor, Sector 2, Noida-201301 UP, India

  • References

  • 1 Honnegowda TM, Kumar P, Udupa EG, Kumar S, Kumar U, Rao P. Role of angiogenesis and angiogenic factors in acute and chronic wound healing. Plast Aesthet Res 2015; 2: 243-249
  • 2 Auerbach R, Auerbach W, Polakowski I. Assays for angiogenesis: a review. Pharmacol Ther 1991; 51 (01) 1-11
  • 3 Adair TH, Montani JP. Angiogenesis. In: Granger DN, Granger JP. eds. Colloquium Series on Integrated Systems Physiology: From Molecule to Function. San Rafael, CA: Morgan & Claypool Life Sciences; 2010: 1-84
  • 4 Wang P, Zhu S, Yuan C, Wang L, Xu J, Liu Z. Shear stress promotes differentiation of stem cells from human exfoliated deciduous teeth into endothelial cells via the downstream pathway of VEGF-Notch signaling. Int J Mol Med 2018; 42 (04) 1827-1836
  • 5 King A, Balaji S, Keswani SG, Crombleholme TM. The role of stem cells in wound angiogenesis. Adv Wound Care (New Rochelle) 2014; 3 (10) 614-625
  • 6 Bindal P, Gnanasegaran N, Bindal U. et al. Angiogenic effect of platelet-rich concentrates on dental pulp stem cells in inflamed microenvironment. Clin Oral Investig 2019; 23 (10) 3821-3831
  • 7 Chakraborty S, Ponrasu T, Chandel S, Dixit M, Muthuvijayan V. Reduced graphene oxide-loaded nanocomposite scaffolds for enhancing angiogenesis in tissue engineering applications. R Soc Open Sci 2018; 5 (05) 172017
  • 8 Kerkis I, Kerkis A, Dozortsev D. et al. Isolation and characterization of a population of immature dental pulp stem cells expressing OCT-4 and other embryonic stem cell markers. Cells Tissues Organs 2006; 184 (3–4): 105-116
  • 9 Miran S, Mitsiadis TA, Pagella P. Innovative dental stem cell-based research approaches: the future of dentistry. Stem Cells Int 2016; 2016: 7231038
  • 10 Li J, Zhu Y, Li N. et al. Upregulation of ETV2 expression promotes endothelial differentiation of human dental pulp stem cells. Cell Transplant 2021; 30: 963689720978739
  • 11 Boreak N, Khayrat NMA, Shami AO. et al. Metformin pre-conditioning enhances the angiogenic ability of the secretome of dental pulp stem cells. Saudi Pharm J 2021; 29 (08) 908-913
  • 12 Li Y, Zhang Y, Wang H. et al. Dental pulp mesenchymal stem cells attenuate limb ischemia via promoting capillary proliferation and collateral development in a preclinical model. Stem Cells Int 2021; 2021: 5585255
  • 13 Li M, Wang Q, Han Q. et al. Novel molecule Nell-1 promotes the angiogenic differentiation of dental pulp stem cells. Front Physiol 2021; 12: 703593
  • 14 Alghutaimel H, Yang X, Drummond B, Nazzal H, Duggal M, Raïf E. Investigating the vascularization capacity of a decellularized dental pulp matrix seeded with human dental pulp stem cells: in vitro and preliminary in vivo evaluations. Int Endod J 2021; 54 (08) 1300-1316
  • 15 Zhou H, Li X, Wu RX. et al. Periodontitis-compromised dental pulp stem cells secrete extracellular vesicles carrying miRNA-378a promote local angiogenesis by targeting Sufu to activate the Hedgehog/Gli1 signalling. Cell Prolif 2021; 54 (05) e13026
  • 16 Huang X, Qiu W, Pan Y. et al. Exosomes from LPS-stimulated hDPSCs activated the angiogenic potential of HUVECs in vitro . Stem Cells Int 2021; 2021: 6685307
  • 17 Afami ME, El Karim I, About I, Coulter SM, Laverty G, Lundy FT. Ultrashort peptide hydrogels display antimicrobial activity and enhance angiogenic growth factor release by dental pulp stem/stromal cells. Materials (Basel) 2021; 14 (09) 2237
  • 18 Liao ZH, Zhu HQ, Chen YY. et al. The epigallocatechin gallate derivative Y6 inhibits human hepatocellular carcinoma by inhibiting angiogenesis in MAPK/ERK1/2 and PI3K/AKT/ HIF-1α/VEGF dependent pathways. J Ethnopharmacol 2020; 259: 112852
  • 19 He Y, Cao Y, Xiang Y. et al. An evaluation of norspermidine on anti-fungal effect on mature Candida albicans biofilms and angiogenesis potential of dental pulp stem cells. Front Bioeng Biotechnol 2020; 8: 948
  • 20 Guo S, Redenski I, Landau S, Szklanny A, Merdler U, Levenberg S. Prevascularized scaffolds bearing human dental pulp stem cells for treating complete spinal cord injury. Adv Healthc Mater 2020; 9 (20) e2000974
  • 21 Luzuriaga J, Irurzun J, Irastorza I, Unda F, Ibarretxe G, Pineda JR. Vasculogenesis from human dental pulp stem cells grown in matrigel with fully defined serum-free culture media. Biomedicines 2020; 8 (11) 483
  • 22 Merckx G, Hosseinkhani B, Kuypers S. et al. Angiogenic effects of human dental pulp and bone marrow-derived mesenchymal stromal cells and their extracellular vesicles. Cells 2020; 9 (02) 312
  • 23 Caseiro AR, Santos Pedrosa S, Ivanova G. et al. Mesenchymal Stem/ Stromal Cells metabolomic and bioactive factors profiles: a comparative analysis on the umbilical cord and dental pulp derived stem/stromal cells secretome. PLoS One 2019; 14 (11) e0221378
  • 24 Makino E, Nakamura N, Miyabe M. et al. Conditioned media from dental pulp stem cells improved diabetic polyneuropathy through anti-inflammatory, neuroprotective and angiogenic actions: cell-free regenerative medicine for diabetic polyneuropathy. J Diabetes Investig 2019; 10 (05) 1199-1208
  • 25 Chen Y, Li X, Wu J, Lu W, Xu W, Wu B. Dental pulp stem cells from human teeth with deep caries displayed an enhanced angiogenesis potential in vitro . J Dent Sci 2021; 16 (01) 318-326
  • 26 Li J, Rao Z, Zhao Y. et al. A decellularized matrix hydrogel derived from human dental pulp promotes dental pulp stem cell proliferation, migration, and induced multidirectional differentiation in vitro . J Endod 2020; 46 (10) 1438-1447.e5
  • 27 Wang D, Lyu Y, Yang Y. et al. Schwann cell-derived EVs facilitate dental pulp regeneration through endogenous stem cell recruitment via SDF-1/CXCR4 axis. Acta Biomater 2022; 140: 610-624
  • 28 Zhou J, Sun C. SENP1/HIF-1α axis works in angiogenesis of human dental pulp stem cells. J Biochem Mol Toxicol 2020; 34 (03) e22436
  • 29 Qu C, Brohlin M, Kingham PJ, Kelk P. Evaluation of growth, stemness, and angiogenic properties of dental pulp stem cells cultured in cGMP xeno-/serum-free medium. Cell Tissue Res 2020; 380 (01) 93-105
  • 30 Zhu L, Dissanayaka WL, Zhang C. Dental pulp stem cells overexpressing stromal-derived factor-1α and vascular endothelial growth factor in dental pulp regeneration. Clin Oral Investig 2019; 23 (05) 2497-2509
  • 31 Li J, Diao S, Yang H, Cao Y, Du J, Yang D. IGFBP5 promotes angiogenic and neurogenic differentiation potential of dental pulp stem cells. Dev Growth Differ 2019; 61 (09) 457-465
  • 32 Lu W, Xu W, Li J, Chen Y, Pan Y, Wu B. Effects of vascular endothelial growth factor and insulin growth factor–1 on proliferation, migration, osteogenesis and vascularization of human carious dental pulp stem cells. Mol Med Rep 2019; 20 (04) 3924-3932
  • 33 Youssef AR, Emara R, Taher MM. et al. Effects of mineral trioxide aggregate, calcium hydroxide, Biodentine and Emdogain on osteogenesis, Odontogenesis, angiogenesis and cell viability of dental pulp stem cells. BMC Oral Health 2019; 19 (01) 133
  • 34 Rapino M, Di Valerio V, Zara S. et al. Chitlac-coated thermosets enhance osteogenesis and angiogenesis in a co-culture of dental pulp stem cells and endothelial cells. Nanomaterials (Basel) 2019; 9 (07) 928
  • 35 Dubey N, Xu J, Zhang Z, Nör JE, Bottino MC. Comparative evaluation of the cytotoxic and angiogenic effects of minocycline and clindamycin: an in vitro study. J Endod 2019; 45 (07) 882-889
  • 36 Delle Monache S, Martellucci S, Clementi L. et al. In vitro conditioning determines the capacity of dental pulp stem cells to function as pericyte-like cells. Stem Cells Dev 2019; 28 (10) 695-706
  • 37 Gong T, Xu J, Heng B. et al. EphrinB2/EphB4 signaling regulates DPSCs to induce sprouting angiogenesis of endothelial cells. J Dent Res 2019; 98 (07) 803-812
  • 38 Schertl P, Volk J, Perduns R. et al. Impaired angiogenic differentiation of dental pulp stem cells during exposure to the resinous monomer triethylene glycol dimethacrylate. Dent Mater 2019; 35 (01) 144-155
  • 39 Luzuriaga J, Pastor-Alonso O, Encinas JM, Unda F, Ibarretxe G, Pineda JR. Human dental pulp stem cells grown in neurogenic media differentiate into endothelial cells and promote neovasculogenesis in the mouse brain. Front Physiol 2019; 10: 347
  • 40 Zou T, Jiang S, Dissanayaka WL. et al. Sema4D/PlexinB1 promotes endothelial differentiation of dental pulp stem cells via activation of AKT and ERK1/2 signaling. J Cell Biochem 2019; 120 (08) 13614-13624
  • 41 Jin R, Song G, Chai J, Gou X, Yuan G, Chen Z. Effects of concentrated growth factor on proliferation, migration, and differentiation of human dental pulp stem cells in vitro . J Tissue Eng 2018; 9: 2041731418817505
  • 42 Gharaei MA, Xue Y, Mustafa K, Lie SA, Fristad I. Human dental pulp stromal cell conditioned medium alters endothelial cell behavior. Stem Cell Res Ther 2018; 9 (01) 69
  • 43 Dou L, Yan Q, Liang P, Zhou P, Zhang Y, Ji P. iTRAQ-based proteomic analysis exploring the influence of hypoxia on the proteome of dental pulp stem cells under 3D culture. Proteomics 2018; 18 (3–4): 1700215
  • 44 Aksel H, Öztürk Ş, Serper A, Ulubayram K. VEGF/BMP-2 loaded three-dimensional model for enhanced angiogenic and odontogenic potential of dental pulp stem cells. Int Endod J 2018; 51 (04) 420-430
  • 45 Lambrichts I, Driesen RB, Dillen Y. et al. Dental pulp stem cells: their potential in reinnervation and angiogenesis by using scaffolds. J Endod 2017; 43 (9S): S12-S16
  • 46 Silva GO, Zhang Z, Cucco C, Oh M, Camargo CHR, Nör JE. Lipoprotein receptor–related protein 6 signaling is necessary for vasculogenic differentiation of human dental pulp stem cells. J Endod 2017; 43 (9S): S25-S30
  • 47 Aksel H, Huang GT. Human and swine dental pulp stem cells form a vascularlike network after angiogenic differentiation in comparison with endothelial cells: a quantitative analysis. J Endod 2017; 43 (04) 588-595
  • 48 Zou T, Dissanayaka WL, Jiang S. et al. Semaphorin 4D enhances angiogenic potential and suppresses osteo-/odontogenic differentiation of human dental pulp stem cells. J Endod 2017; 43 (02) 297-305
  • 49 Nam H, Kim GH, Bae YK. et al. Angiogenic capacity of dental pulp stem cell regulated by SDF-1 α-CXCR4 axis. Stem Cells Int 2017; 2017: 8085462
  • 50 Lee SI, Lee ES, El-Fiqi A, Lee SY, Eun-Cheol Kim Kim HW. Stimulation of odontogenesis and angiogenesis via bioactive nanocomposite calcium phosphate cements through integrin and VEGF signaling pathways. J Biomed Nanotechnol 2016; 12 (05) 1048-1062
  • 51 Lee SI, Kim SY, Park KR, Kim EC. Baicalein promotes angiogenesis and odontoblastic differentiation via the BMP and Wnt pathways in human dental pulp cells. Am J Chin Med 2016; 44 (07) 1457-1472
  • 52 Spina A, Montella R, Liccardo D. et al. NZ-GMP approved serum improve hDPSC osteogenic commitment and increase angiogenic factor expression. Front Physiol 2016; 7: 354
  • 53 Kuang R, Zhang Z, Jin X. et al. Nanofibrous spongy microspheres for the delivery of hypoxia-primed human dental pulp stem cells to regenerate vascularized dental pulp. Acta Biomater 2016; 33: 225-234
  • 54 Shen C, Li L, Feng T. et al. Dental pulp stem cells derived conditioned medium promotes angiogenesis in hindlimb ischemia. J Tissue Eng Regen Med 2015; 12 (01) 59-68
  • 55 Dissanayaka WL, Zhu L, Hargreaves KM, Jin L, Zhang C. In vitro analysis of scaffold-free prevascularized microtissue spheroids containing human dental pulp cells and endothelial cells. J Endod 2015; 41 (05) 663-670
  • 56 Boyle M, Chun C, Strojny C. et al. Chronic inflammation and angiogenic signaling axis impairs differentiation of dental-pulp stem cells. PLoS One 2014; 9 (11) e113419
  • 57 Liu W, Gong Q, Ling J, Zhang W, Liu Z, Quan J. Role of miR-424 on angiogenic potential in human dental pulp cells. J Endod 2014; 40 (01) 76-82
  • 58 Bronckaers A, Hilkens P, Fanton Y. et al. Angiogenic properties of human dental pulp stem cells. PLoS One 2013; 8 (08) e71104
  • 59 Janebodin K, Zeng Y, Buranaphatthana W, Ieronimakis N, Reyes M. VEGFR2-dependent angiogenic capacity of pericyte-like dental pulp stem cells. J Dent Res 2013; 92 (06) 524-531
  • 60 Ishizaka R, Hayashi Y, Iohara K. et al. Stimulation of angiogenesis, neurogenesis and regeneration by side population cells from dental pulp. Biomaterials 2013; 34 (08) 1888-1897
  • 61 Dissanayaka WL, Zhan X, Zhang C, Hargreaves KM, Jin L, Tong EH. Coculture of dental pulp stem cells with endothelial cells enhances osteo-/odontogenic and angiogenic potential in vitro . J Endod 2012; 38 (04) 454-463
  • 62 Iohara K, Zheng L, Wake H. et al. A novel stem cell source for vasculogenesis in ischemia: subfraction of side population cells from dental pulp. Stem Cells 2008; 26 (09) 2408-2418
  • 63 Wu M, Liu X, Li Z. et al. SHED aggregate exosomes shuttled miR-26a promote angiogenesis in pulp regeneration via TGF-β/SMAD2/3 signalling. Cell Prolif 2021; 54 (07) e13074
  • 64 Han Y, Chen Q, Zhang L, Dissanayaka WL. Indispensable role of HIF-1α signaling in post-implantation survival and angio-/vasculogenic properties of SHED. Front Cell Dev Biol 2021; 9: 655073
  • 65 Zaw SYM, Kaneko T, Zaw ZCT. et al. Crosstalk between dental pulp stem cells and endothelial cells augments angiogenic factor expression. Oral Dis 2020; 26 (06) 1275-1283
  • 66 Atlas Y, Gorin C, Novais A. et al. Microvascular maturation by mesenchymal stem cells in vitro improves blood perfusion in implanted tissue constructs. Biomaterials 2021; 268: 120594
  • 67 Guo H, Zhao W, Liu A. et al. SHED promote angiogenesis in stem cell-mediated dental pulp regeneration. Biochem Biophys Res Commun 2020; 529 (04) 1158-1164
  • 68 Gong T, Heng BC, Xu J. et al. Decellularized extracellular matrix of human umbilical vein endothelial cells promotes endothelial differentiation of stem cells from exfoliated deciduous teeth. J Biomed Mater Res A 2017; 105 (04) 1083-1093
  • 69 Kim JH, Kim GH, Kim JW. et al. In vivo angiogenic capacity of stem cells from human exfoliated deciduous teeth with human umbilical vein endothelial cells. Mol Cells 2016; 39 (11) 790-796
  • 70 Gorin C, Rochefort GY, Bascetin R. et al. Priming dental pulp stem cells with fibroblast growth factor-2 increases angiogenesis of implanted tissue-engineered constructs through hepatocyte growth factor and vascular endothelial growth factor secretion. Stem Cells Transl Med 2016; 5 (03) 392-404
  • 71 Bento LW, Zhang Z, Imai A. et al. Endothelial differentiation of SHED requires MEK1/ERK signaling. J Dent Res 2013; 92 (01) 51-57
  • 72 Iwasaki K, Akazawa K, Nagata M. et al. Angiogenic effects of secreted factors from periodontal ligament stem cells. Dent J 2021; 9 (01) 9
  • 73 Zhang Z, Shuai Y, Zhou F. et al. PDLSCs regulate angiogenesis of periodontal ligaments via VEGF transferred by exosomes in periodontitis. Int J Med Sci 2020; 17 (05) 558-567
  • 74 Diomede F, Marconi GD, Cavalcanti MFXB. et al. VEGF/VEGF-R/RUNX2 upregulation in human periodontal ligament stem cells seeded on dual acid etched titanium disk. Materials (Basel) 2020; 13 (03) 706
  • 75 Marconi GD, Diomede F, Pizzicannella J. et al. Enhanced VEGF/VEGF-R and RUNX2 expression in human periodontal ligament stem cells cultured on sandblasted/etched titanium disk. Front Cell Dev Biol 2020; 8: 315
  • 76 Kim HJ, Yoo JH, Choi Y, Joo JY, Lee JY, Kim HJ. Assessing the effects of cyclosporine A on the osteoblastogenesis, osteoclastogenesis, and angiogenesis mediated by human periodontal ligament stem cells. J Periodontol 2020; 91 (06) 836-848
  • 77 Iwasaki K, Nagata M, Akazawa K, Watabe T, Morita I. Changes in characteristics of periodontal ligament stem cells in spheroid culture. J Periodontal Res 2019; 54 (04) 364-373
  • 78 Jearanaiphaisarn T, Sanharati T, Pavasant P, Nakalekha Limjeerajarus C. The effect of iloprost on cell proliferation and angiogenesis-related gene expression in human periodontal ligament cells. Odontology 2018; 106 (01) 11-18
  • 79 Wei W, An Y, An Y, Fei D, Wang Q. Activation of autophagy in periodontal ligament mesenchymal stem cells promotes angiogenesis in periodontitis. J Periodontol 2018; 89 (06) 718-727
  • 80 Bae YK, Kim GH, Lee JC. et al. The significance of SDF-1α-CXCR4 axis in in vivo angiogenic ability of human periodontal ligament stem cells. Mol Cells 2017; 40 (06) 386-392
  • 81 Yi B, Ding T, Jiang S. et al. Conversion of stem cells from apical papilla into endothelial cells by small molecules and growth factors. Stem Cell Res Ther 2021; 12 (01) 266
  • 82 Liu J, Zou T, Yao Q, Zhang Y, Zhao Y, Zhang C. Hypoxia-mimicking cobalt-doped multi-walled carbon nanotube nanocomposites enhance the angiogenic capacity of stem cells from apical papilla. Mater Sci Eng C 2021; 120: 111797
  • 83 Yu S, Zhao Y, Fang TJ, Ge L. Effect of the soluble factors released by dental apical papilla-derived stem cells on the osteo/odontogenic, angiogenic, and neurogenic differentiation of dental pulp cells. Stem Cells Dev 2020; 29 (12) 795-805
  • 84 Yuan C, Wang P, Zhu S. et al. Overexpression of ephrinB2 in stem cells from apical papilla accelerates angiogenesis. Oral Dis 2019; 25 (03) 848-859
  • 85 Koutsoumparis A, Vassili A, Bakopoulou A, Ziouta A, Tsiftsoglou AS. Erythropoietin (rhEPOa) promotes endothelial transdifferentiation of stem cells of the apical papilla (SCAP). Arch Oral Biol 2018; 96: 96-103
  • 86 Yadlapati M, Biguetti C, Cavalla F. et al. Characterization of a vascular endothelial growth factor–loaded bioresorbable delivery system for pulp regeneration. J Endod 2017; 43 (01) 77-83
  • 87 Yuan C, Wang P, Zhu S. et al. EphrinB2 stabilizes vascularlike structures generated by endothelial cells and stem cells from apical papilla. J Endod 2016; 42 (09) 1362-1370
  • 88 Peters OA, Galicia J, Arias A, Tolar M, Ng E, Shin SJ. Effects of two calcium silicate cements on cell viability, angiogenic growth factor release and related gene expression in stem cells from the apical papilla. Int Endod J 2016; 49 (12) 1132-1140
  • 89 Bakopoulou A, Kritis A, Andreadis D. et al. Angiogenic potential and secretome of human apical papilla mesenchymal stem cells in various stress microenvironments. Stem Cells Dev 2015; 24 (21) 2496-2512
  • 90 Yuan C, Wang P, Zhu L. et al. Coculture of stem cells from apical papilla and human umbilical vein endothelial cell under hypoxia increases the formation of three-dimensional vessel-like structures in vitro . Tissue Eng Part A 2015; 21 (5–6): 1163-1172
  • 91 Jin S, Yang C, Huang J. et al. Conditioned medium derived from FGF-2-modified GMSCs enhances migration and angiogenesis of human umbilical vein endothelial cells. Stem Cell Res Ther 2020; 11 (01) 68
  • 92 Zhu SY, Yuan CY, Lin YF. et al. Stem cells from human exfoliated deciduous teeth (SHEDs) and dental pulp stem cells (DPSCs) display a similar profile with pericytes. Stem Cells Int 2021; 2021: 8859902
  • 93 Xie J, Zhao YM, Rao NQ. et al. Comparative study of differentiation potential of mesenchymal stem cells derived from orofacial system into vascular endothelial cells. Beijing Da Xue Xue Bao Yi Xue Ban 2019; 51 (05) 900-906
  • 94 Angelopoulos I, Brizuela C, Khoury M. Gingival mesenchymal stem cells outperform haploidentical dental pulp-derived mesenchymal stem cells in proliferation rate, migration ability, and angiogenic potential. Cell Transplant 2018; 27 (06) 967-978
  • 95 Xu JG, Gong T, Wang YY. et al. Inhibition of TGF-β signaling in SHED enhances endothelial differentiation. J Dent Res 2018; 97 (02) 218-225
  • 96 Osman A, Gnanasegaran N, Govindasamy V. et al. Basal expression of growth-factor-associated genes in periodontal ligament stem cells reveals multiple distinctive pathways. Int Endod J 2014; 47 (07) 639-651
  • 97 Zhang Z, Oh M, Sasaki JI, Nör JE. Inverse and reciprocal regulation of p53/p21 and Bmi-1 modulates vasculogenic differentiation of dental pulp stem cells. Cell Death Dis 2021; 12 (07) 644
  • 98 Olcay K, Taşli PN, Güven EP. et al. Effect of a novel bioceramic root canal sealer on the angiogenesis-enhancing potential of assorted human odontogenic stem cells compared with principal tricalcium silicate-based cements. J Appl Oral Sci 2020; 28: e20190215
  • 99 Hilkens P, Bronckaers A, Ratajczak J, Gervois P, Wolfs E, Lambrichts I. The angiogenic potential of DPSCs and SCAPs in an in vivo model of dental pulp regeneration. Stem Cells Int 2017; 2017: 2582080
  • 100 Zhang Z, Nör F, Oh M, Cucco C, Shi S, Nör JE. Wnt/β-catenin signaling determines the vasculogenic fate of postnatal mesenchymal stem cells. Stem Cells 2016; 34 (06) 1576-1587
  • 101 Hilkens P, Fanton Y, Martens W. et al. Pro-angiogenic impact of dental stem cells in vitro and in vivo . Stem Cell Res (Amst) 2014; 12 (03) 778-790
  • 102 Gandia C, Armiñan A, García-Verdugo JM. et al. Human dental pulp stem cells improve left ventricular function, induce angiogenesis, and reduce infarct size in rats with acute myocardial infarction. Stem Cells 2008; 26 (03) 638-645
  • 103 Zeidán-Chuliá F, Noda M. “Opening” the mesenchymal stem cell tool box. Eur J Dent 2009; 3 (03) 240-249
  • 104 Fiorillo L. Fi-index: a new method to evaluate authors Hirsch-index reliability. Publ Res Q 2022; 38: 465-474
  • 105 Fiorillo L, Cicciù M. The use of Fi-index tool to assess per-manuscript self-citations. Publ Res Q 2022; 38: 684-692

Zoom Image
Fig. 1 Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) chart illustrating the research methodology used in the review.
Zoom Image
Fig. 2 Graphical representation of the source-wise articles included in the review. DPSC, dental pulp stem cell; GMSC, gingival mesenchymal stem cell; PDLSC, periodontal ligament–derived mesenchymal stem cell; SCAP, stem cells from apical papilla; SHED, stem cells from human exfoliated deciduous teeth.
Zoom Image
Fig. 3 Summary of the meta-analysis assessing the effect of DPMSCs on the tubular length in an in vitro Matrigel assay showing a positive correlation of the co-culture of HUVEC and DPMSCs with the tubule length formation, which was statistically significant (p = 0.04). CI, confidence interval; DPMSCs, dental pulp–derived mesenchymal stem cells; DPSC, dental pulp stem cell; HUVEC, human umbilical vein endothelial cell; SD, standard deviation.
Zoom Image
Fig. 4 Summary of the meta-analysis assessing the effect of SCAP on the tubular length in an in vitro Matrigel assay showing a positive correlation of the co-culture of HUVEC and SCAPs with the tubule length formation, which was not statistically significant (p = 0.16). CI, confidence interval; HUVEC, human umbilical vein endothelial cell; SCAP, stem cells from apical papilla; SD, standard deviation.
Zoom Image
Fig. 5 Summary of the meta-analysis assessing the effect of SCAP on the total branching points in an in vitro Matrigel assay showing a positive correlation of the co-culture of HUVEC and SCAPs with the total branching point number, which was not statistically significant (p = 0.14). CI, confidence interval; HUVEC, human umbilical vein endothelial cell; SCAP, stem cells from apical papilla; SD, standard deviation.
Zoom Image
Fig. 6 Graphical representation of Egger's regression test.