Abstract

Chronic cardiometabolic assaults during type 2 diabetes (T2D) and obesity induce a progenitor cell imbalance in the circulation characterized by overproduction and release of pro-inflammatory monocytes and granulocytes from the bone marrow alongside aberrant differentiation and mobilization of pro-vascular progenitor cells that generate downstream progeny for the coordination of blood vessel repair. This imbalance can be detected in the peripheral blood of individuals with established T2D and severe obesity using multiparametric flow cytometry analyses to discern pro-inflammatory vs. pro-angiogenic progenitor cell subsets identified by high aldehyde dehydrogenase activity, a conserved progenitor cell protective function, combined with lineage-restricted cell surface marker analyses. Recent evidence suggests that progenitor cell imbalance can be reversed by treatment with pharmacological agents or surgical interventions that reduce hyperglycaemia or excess adiposity. In this state-of-the-art review, we present current strategies to assess the progression of pro-vascular regenerative cell depletion in peripheral blood samples of individuals with T2D and obesity and we summarize novel clinical data that intervention using sodium-glucose co-transporter 2 inhibition or gastric bypass surgery can efficiently restore cell-mediated vascular repair mechanisms associated with profound cardiovascular benefits in recent outcome trials. Collectively, this thesis generates a compelling argument for early intervention using current pharmacological agents to prevent or restore imbalanced circulating progenitor content and maintain vascular regenerative cell trafficking to sites of ischaemic damage. This conceptual advancement may lead to the design of novel therapeutic approaches to prevent or reverse the devastating cardiovascular comorbidities currently associated with T2D and obesity.

During cardiometabolic based chronic disease, dysglycemia and excess adipocity induce a differentiate imbalance in the bone marrow that contribute to overproduction of inflammatory cells and impaired mobilization of progenitor cells that mediate endogenous mechanisms for vessel repair and regeneration.
Graphical Abstract

During cardiometabolic based chronic disease, dysglycemia and excess adipocity induce a differentiate imbalance in the bone marrow that contribute to overproduction of inflammatory cells and impaired mobilization of progenitor cells that mediate endogenous mechanisms for vessel repair and regeneration.

Introduction to chronic cardiometabolic disease

Link between chronic cardiometabolic disease and cardiovascular comorbidities

The clinical outcomes of ischaemic cardiovascular diseases (CVD), including coronary and peripheral artery disease, myocardial infarction, heart failure and stroke, are often considered unavoidable complications that compromises quality of life in patients with type 2 diabetes (T2D) and obesity. CVD was responsible for nearly 18 million global deaths in 2015,1  ,  2 and collectively CVD remains the leading cause of death worldwide.3  ,  4 It is estimated that 420 million individuals are living with diabetes and 2 billion individuals are considered overweight, 650 million of which are affected by obesity [body mass index (BMI) >30 kg/m2]. Due to the increased prevalence of T2D and obesity in an ageing population worldwide, and despite improved pharmacologic and surgical intervention, the incidence of CVD is expected to continue to rise over the next decade.5 The global financial burden of treating diabetes was estimated at $1.31 trillion in 2015,6 and CVD comorbidities were responsible for nearly one-third ($437 billion) of the cost of therapy.7 Thus, there exists a compelling need to better understand the pathophysiological interrelationship between T2D, obesity and the subsequent development of CVD.

In a recent review, Mechanick et al.8 conceptualized that ischaemic CVD represents the endpoint of an accumulation of clinical risk factors including hyperglycaemia and insulin resistance during T2D and the elevation of systemic inflammation during obesity. Because T2D and obesity are often highly entangled, comorbid states, chronic dysglycaemia, and increased adiposity can be considered primary drivers of vascular damage that overwhelm endogenous vessel repair mechanisms.9–13 This unmanaged cycle of excessive vascular damage combined with gradually compromised vessel regenerative response can intensify atherosclerotic plaque instability and elevate the risk of experiencing ischaemic macrovascular events,14 including myocardial infarction and stroke. Therefore, strategies to stimulate the recovery of endogenous blood vessel repair represent a novel target to prevent and treat CVD progression15 (See Graphical Abstract).

Progenitor cells that mediate blood vessel repair

Endogenous processes for blood vessel repair and regeneration

In response to tissue hypoxia or ischaemic injury, bone marrow (BM)-derived haematopoietic progenitor cells (HPC) and rare endothelial progenitor cells (EPC) are stimulated by neurohormonal signals and chemokines in the BM to mobilize into the peripheral circulation. As such, endogenous vascular regeneration is bolstered by haemangiogenic precursor cells and their downstream progeny that recruit to sites of vessel damage. Blood vessel repair and regeneration is typically classified into THREE overlapping multicellular processes: (i) vasculogenesis, the de novo synthesis of new blood vessels by circulating EPC first identified by Asahara and Isner;16  ,  17 (ii) angiogenesis, the sprouting of new blood vessels from pre-existing vessels stimulated by angiocrine secretion from endothelial cells and circulating haematopoietic cells; and (iii) arteriogenesis, the remodelling of pre-existing collateral vessels by monocytes that polarize into regenerative macrophages in peripheral tissues.18  ,  19 These complementary processes occur simultaneously to synergize blood vessel regeneration, repair and anastomoses.20 Thus, healthy human blood contains circulating BM-derived progenitor cells that contribute to the maintenance of blood vessel homeostasis throughout life. However, in individuals with T2D and obesity, hyperglycaemia and adiposity elevate oxidative stress and inflammation to generate a ‘perfect storm’ that alters progenitor cell differentiation upsetting the execution of blood vessel repair mechanisms.

Circulating CD34+VEGFR-2+ cells are decreased during type 2 diabetes and obesity

The seminal discovery by Asahara et al.  16 and Isner et al.  17 first defined EPC as highly proliferative CD34+ cells that could be mobilized from the BM and integrate into vascular structures in vivo. Because mature endothelial cells within blood vessels are remarkably quiescent with an estimated half-life of ∼3 years, Asahara et al.  21 first suggested that post-natal vasculogenesis, a process initially thought to be restricted to the developing embryo, could be mediated in adult organisms after transplantation or mobilization of EPC from the BM. Later, cell surface markers such as CD133 and vascular endothelial growth factor receptor-2 (VEGFR-2) were added to better define the putative EPC population. In 2001, Vasa et al.  22 were the first to report that the number and migratory function of EPC (identified as CD34+/VEGFR-2+ cells) was severely reduced in patients with coronary artery disease (CAD). Individuals with CAD had ∼50% fewer circulating CD34+/VEGFR-2+ cells compared to healthy controls. Furthermore, common risk factors associated with CAD correlated with decreased circulating CD34+/VEGFR-2+ content. Smoking was the most significant predictor for EPC depletion, alongside hypertension, serum low-density lipoprotein (LDL) cholesterol, age and previous family history of CAD.22 Reciprocally, Werner et al.23 observed that in subjects with CAD, individuals in the top tertile for circulating CD34+/VEGFR-2+ cells demonstrated a reduced risk of major cardiovascular events. Zafar et al. recently enrolled a human cohort with variable CVD risk and demonstrated an association between different progenitor cell subsets within indices of T2D. Subjects with a lower frequency of CD34+/CD133+ cells had a significantly higher incidence of T2D, higher glycated haemoglobin (HbA1c), higher CVD risk scores, and impaired vascular function.24 Circulating CD34+/CD133+ cells from patients with T2D demonstrated reduced CXCR4 and VEGFR-2 expression, suggesting a homing deficit to areas of ischaemia. Surprisingly, existing CD34+/CD133+ cells in the high-risk group compensated with increased expression of angiopoietins, VEGF-A, PlGF, HGF, EPO, and MMP-9 compared to the lower risk cohort. Thus, reduced circulating progenitor cell content and recruitment to sites of vascular injury were implicated with T2D pathology without an observed deficit in angiogenic cytokine expression.

In a series of studies, Fadini et al.  25  ,  26 sought to determine if hyperglycaemia resulted in EPC functional impairment. In hyperglycaemic mice with hind-limb ischaemic injury, CD34+/VEGFR2+ cells were ineffectively mobilized into the periphery compared to normoglycaemic mice. Insulin administration to promote glycaemic control induced partial recovery of circulating CD34+/VEGFR2+ cells. In individuals with T2D, peripheral artery disease was independently associated with reduced circulating CD34+/VEGFR2+ content,25 and CD34+/VEGFR2+ isolated from individuals with peripheral artery disease demonstrated reduced ability to form endothelial cell colonies in vitro. In contrast, a mild elevation in circulating CD34+ cells in healthy individuals was associated with increasing adiposity and insulin resistance.27 Collectively, these studies established CD34, CD133, and VEGFR-2 as biomarkers to document changes in circulating progenitor cell content during T2D and obesity. However, the identity and function of ‘true’ EPC remained elusive and soon met controversy surrounding the physical contribution of BM-derived progenitor cells into the growing vasculature in healthy and diseased conditions. For a comprehensive review on circulating stem cells and cardiovascular outcomes refer to Fadini et al.28

The endothelial progenitor cell controversy: CD34+/VEGFR-2+ cells primarily represent progenitor cells of the myeloid lineage

Haematopoiesis is highly integrated with blood vessel repair and maintenance. HPC differentiate into myeloid progeny that give rise to reticulated erythrocytes, monocytes and granulocytes, and megakaryocytes, essential for the transport of oxygen, innate immune surveillance, and platelet production, respectively.29 In addition, primitive HPC and monocytes also possess potent pro-angiogenic secretory function and stimulate microvascular capillary formation to recover perfusion in mice with hindlimb ischaemia.30 Indeed, mouse embryos deficient in acute myeloid leukaemia 1 protein (AML1), a transcription factor necessary for myeloid cell differentiation, were incapable of stimulating angiogenesis resulting in embryonic lethality.30 Notably, embryo lethality could be rescued by incubation with BM-derived HPC, suggesting the profound role of myeloid progenitor cell secretions to stimulate blood vessel regeneration throughout adult life.

Canonically, classification of EPC based on CD34 or VEGFR-2 expression has generated confusion in the field since myeloid HPC also express these markers. In seminal findings by the Dimmeler group, Urbich et al.31 demonstrated that CD34+/VEGFR-2+ cells, referred to as EPC in previous work, consisted primarily of cells from the haematopoietic lineage. Termed circulating angiogenic cells (CAC), endothelial-cell-like cultures could be established from CD14+ and CD14- mononuclear cells, both cell types demonstrated lectin uptake in vitro, and stimulated neovascularization after transplantation into mouse models of hind-limb ischaemia. The CD14+ cells were comprised of a mixture of monocytes, macrophages, and T lymphocytes, that did not produce endothelial cell progeny, but contributed to capillary formation with potent angiocrine function. Yoder et al. first delineated the functional differences between ‘true EPC’ termed endothelial colony-forming cells (ECFC) and CAC based on fundamental stem and progenitor cell principles.32 CAC formed early outgrowth (7–10 days) colonies in culture, co-expressed endothelial (CD34, VEGFR-2) and haematopoietic cell markers (CD45+/CD14+), and secreted cytokines to promote tip cell formation during angiogenic vessel sprouting but did not integrate into vessels in vivo. In contrast, ECFC were exclusively CD45/CD14, formed late outgrowth (>14 days) endothelial cell colonies in vitro, and could integrate into blood vessels in vivo. Nonetheless, these findings suggested novel roles for CD34+ HPC and CD14+ monocytes to stimulate vessel repair alongside circulating or vessel-resident EPC as a potential source of endothelial cells.

Arteriogenic remodelling is impaired during type 2 diabetes and obesity

Angiogenic and arteriogenic vessel repair mechanisms are distinguished based on distinct physiological drivers. Angiogenesis occurs primarily as a response to hypoxia, driving hypoxia-inducible factor 1α (HIF-1α)-mediated transcription of VEGF and chemokines that stimulate surveillant CAC to recruit to areas of ischaemia and secrete a variety of pro-angiogenic cytokines. In contrast, the predominant process to bypass vessels occluded during arteriogenesis occurs in response to fluid shear stress. Although arteriogenesis is not associated with increases in HIF-1α or VEGF,33 reduced luminal diameter in atherosclerotic vessels increases shear stress on the vascular endothelium, stimulating the secretion of powerful chemokines (such as monocyte chemoattractant protein-1, MCP-1)34 and expression of adhesion molecules (such as integrins) to recruit and adhere to CAC. Risk factors such as smoking35 and T2D36–38 have been shown to impair both angiogenic capillary formation and arteriogenic collateral vessel remodelling. Thus, better understanding of the relationship between circulating monocytes and development of tissue remodelling macrophages that synergize arteriogenic collateral vessel formation represents an ongoing focus to modulate large vessel regeneration.

Monocytes are largely considered precursors to matrix-remodelling macrophages. Within tissues, monocyte populations can adopt pro-inflammatory M1-like or pro-regenerative M2-like fates (further subdivided into M2a, M2b, and M2c groups).39 Notably, this oversimplified description of macrophage subtypes is better described as a continuum of highly plastic cells that play a vital role in arteriogenic tissue remodelling. In concept, M1-like macrophages primarily represent phagocytic cells that secrete pro-inflammatory cytokines such as interleukin (IL)-1β, IL-6, IL-12, and tumor necrosis factor-α and recruit neutrophils that propagate inflammation. Conversely, M2-like polarized macrophages are considered anti-inflammatory and mediate extracellular remodelling via the secretion of growth factors such as transforming growth factor-β (TGF-β) and fibroblast growth factor (FGF). Together, collateral artery activation relies on the recruitment of monocytes that polarize along the M1/M2 continuum to dictate the delicate balance between tissue inflammation vs. regenerative arteriogenesis.

Troidl et al.40 sought to determine the contribution of M1- and M2-polarized macrophages to arteriogenesis using femoral artery ligation in rodent models to induce shear stress. M2-polarized macrophages were scarcely distributed throughout the perivascular space prior to femoral artery ligation. After occlusion, both M1 and M2 polarized macrophage content was increased. M2-like macrophages augmented matrix degradation at the leading edge of remodelling vessels, highlighting a critical role for M2 macrophages to facilitate collateral activation. Takeda et al.41 observed that M2-polarized macrophages also secreted SDF-1 and PDGFB to recruit pericytes involved in blood vessel stabilization. Thus, collateral activation via arteriogenesis represents a novel target for blood vessel repair by altering the balance between M1-like and M2-like macrophage content associated with altered blood flow parameters.

High ALDH activity identifies progenitor cells with potent angiocrine function

To identify blood-derived progenitor cells with vascular regenerative potential, our group and others purify and expand human progenitor cell subsets based on selection for high aldehyde dehydrogenase (ALDH) activity.42–45 ALDH is a intracellular detoxification enzyme highly expressed by progenitor cells from multiple mesodermal lineages that protects differentiating progenitor cells from damage by alkylating agents or oxidative stress. As progenitor cells with high ALDH activity differentiate towards more expendable effector cells, ALDH activity is decreased up to 100-fold. By assessing the distribution of peripheral blood cells with high vs. low ALDH activity (Aldefluor™ assay; StemCell Technologies) by flow cytometry, we have identified a unique functional biomarker for progenitor cells (ALDHhi cells) vs. more differentiated cell progeny (ALDHlow cells). Storms et al.45  ,  46 first identified human BM cells with high ALDH activity as myeloid HPC demonstrating multipotent colony formation in vitro. Because ALDHhiSSClow cells highly co-expressed CD34 (>75%) and CD133 (>55%), BM-derived ALDHhiSSClow cells demonstrated robust haematopoietic repopulation capacity in immunodeficient mice.47  ,  48 In Capoccia et al., we revealed that human BM-derived ALDHhiSSClow cells represented a heterogeneous mixture of progenitor cells with haematopoietic (1 HCFC in 4 cells), endothelial (∼1 in ECFC 1500 cells), and fibroblast (∼1 FCFC in 1500 cells) colony-forming cell activity in vitro. Compared to mature mononuclear cells, purified BM ALDHhiSSClow cells possessed a pro-angiogenic secretory profile and accelerated the recovery from ischaemia when transplanted into immunodeficient mice with unilateral hind-limb ischaemia induced after femoral artery ligation and excision.49 Thus, ALDH activity represented a valuable tool to purify progenitor cells with vessel regenerative capacity based on a tightly conserved progenitor cell function. These findings were immediately translated into a clinical trial by Perin et al.  50 for severe critical limb ischaemia demonstrating that intramuscular implantation of autologous BM ALDHhi cells improved Rutherford category scores and ankle-brachial index at 12 weeks post-transplantation. However, in a study of 82 patients, the PACE trial recently reported that transplantation of autologous ALDHhi cells in patients with intermittent claudication showed no improvement in capillary perfusion compared to placebo, but ALDHhi cell administration was associated with an increase in the number of collateral arteries.51

The assessment of ALDH activity alongside cell granularity (increasing side scatter property) and the co-expression of lineage-specific cell surface markers within peripheral blood of individuals with T2D, has also provided a high-throughput method to compare circulating progenitor cell subsets previously associated with pro-inflammatory vs. pro-angiogenic properties.15 Our rationale was to assess1 circulating ALDHhiSSClow cells, representing uncommitted HPC with a pro-angiogenic secretome,52 alongside primitive cell surface marker (CD34, CD133, and VEGFR-2) co-expression; circulating ALDHhiSSCmid cells, representing monocyte precursors with M1 (CD80) vs. M2 (CD163, CD206) polarization, and ALDHhiSSChi cells, representing granulocytic precursors with pro-inflammatory (CD15, CD16) or pro-angiogenic marker (CD49d) co-expression. Importantly, ALDHlow cells from each SSC classification represented more differentiated cells from each lineage.

Reversal of progenitor cell imbalance during metabolic diseases

Circulating progenitor cell imbalance during type 2 diabetes

Homeostatic vascular remodelling occurs continually throughout growth and ageing. Indeed, the revascularization of damaged or ischaemic tissues represents a central component of all regenerative processes. Derived in the BM and mobilized to areas of hypoxia via the bloodstream, ALDHhiSSClow progenitor cells generate and direct the myriad of effector cells that participate in angiogenic, vasculogenic, and arteriogenic processes. Although human organs possess an intrinsic capacity for vessel regeneration, hyperglycaemia and increasing adiposity during T2D and obesity result in relentless damage that depletes the pro-angiogenic cell reservoir and forces the production of pro-inflammatory granulocyte progeny53 (Figure 1). This circulating progenitor cell imbalance encompasses the depletion or dysfunction of cells associated with vessel repair,54–57 and the overproduction of granulocytes and monocytes that heighten inflammation during T2D and obesity. Thus, progressive vessel damage driven by hyperglycaemia or atherosclerosis is compounded by significant deficits in vessel repair, ultimately elevating the risk of developing ischaemic CVD.58

Circulating Progenitor Cell Imbalance elevates systemic inflammation and reduces circulating cell types that mediate blood vessel repair. Circulating progenitor cell imbalance results in reduced maintenance of ALDHhiSSClow progenitor cells with CD34 and CD133 co-expression and altered haematopoietic differentiation towards the overproduction of pro-inflammatory granulocytes at the expense of vessel regenerative cell types. Altered monocyte differentiation promoted the production of ALDHhiSSCmid monocytes with M1-polarization vs. M2-polarization. Collectively, altered circulating progenitor cell content impairs endogenous cell-mediated blood vessel repair and elevates systemic inflammation during type 2 diabetes and severe obesity.
Figure 1

Circulating Progenitor Cell Imbalance elevates systemic inflammation and reduces circulating cell types that mediate blood vessel repair. Circulating progenitor cell imbalance results in reduced maintenance of ALDHhiSSClow progenitor cells with CD34 and CD133 co-expression and altered haematopoietic differentiation towards the overproduction of pro-inflammatory granulocytes at the expense of vessel regenerative cell types. Altered monocyte differentiation promoted the production of ALDHhiSSCmid monocytes with M1-polarization vs. M2-polarization. Collectively, altered circulating progenitor cell content impairs endogenous cell-mediated blood vessel repair and elevates systemic inflammation during type 2 diabetes and severe obesity.

Because a non-invasive assay was needed to accurately document the circulating progenitor cell imbalance in patients with T2D and obesity, we combined ALDH activity with cell type-specific surface marker analyses to develop a high-throughput flow cytometry platform to quantify lineage-restricted progenitor cell content within peripheral blood samples from patients with T2D compared to sex- and age-matched control subjects.59 Compared to non-diabetic controls, individuals with established T2D for a mean duration of >15 years exhibited: (i) an increased frequency of circulating ALDHhiSSChi granulocyte precursors, (ii) an increased frequency of ALDHhiSSCmid monocytes with M1 polarization, and (iii) depletion of ALDHhiSSClow precursor cells that co-expressed CD34 or CD133 with pro-angiogenic function.47 Notably, the duration of T2D directly correlated with the severity of circulating progenitor cell imbalance. Collectively, these observations further refined the progenitor cell imbalance as a reduction in circulating HPC combined with myelopoietic overproduction of inflammatory monocytes and granulocytes consistent with published mechanistic studies in mouse models of hyperglycaemia60  ,  61 and obesity.62 This novel assessment of circulating ALDHhi progenitor cell subsets represented a promising approach to identify T2D patients with compromised blood vessel repair capacity.

SGLT2 inhibition reversed the progenitor cell imbalance in individuals with T2D

Chronic hyperglycaemia (HbA1c >6.5%) represents an important driver for the development of microvascular risk in patients with T2D. Sodium-glucose co-transporter 2 inhibitors (SGLT2i), including dapagliflozin, canagliflozin, and empagliflozin, represent a novel class of glucose-lowering medications that block the reabsorption of glucose and sodium from the kidney to the bloodstream, increasing urinary glucose excretion and thereby lowering HbA1c levels.63 However, the benefits of SGLT2i may extend beyond glycaemic control. The EMPA-REG OUTCOME trial established that empagliflozin treatment in patients with T2D correlated with significant reductions in major adverse cardiovascular events (MACE) and all-cause mortality.64 However, the mechanisms driving these improved outcomes remain undetermined.

In human BM, hyperglycaemia has been shown to induce premature inflammatory cell differentiation and inhibition of vascular regenerative progenitor cell egression into the periphery65 (Figure 1). Performed as a sub-study of the EMPA-Heart CardioLink-6 trial, we investigated whether administration of empagliflozin could impact the frequencies of circulating ALDHhi progenitor cell subsets in patients with established T2D over a 6-month period.66 Remarkably, individual with T2D administered empagliflozin demonstrated reduced circulating ALDHhiSSChi granulocyte precursor burden, an improved ratio of ALDHhiSSCmid M1- vs. M2-polarized monocytes, and increased ALDHhiSSClow CD34+/CD133+ progenitor cell content compared to subjects receiving standard medical care. In the plasma of subjects administered empagliflozin, we also documented stabilization of NADPH oxidase 1 (NOX1), an enzyme dedicated to the production of reactive oxygen species (ROS), alongside increased catalase, an anti-oxidant that accelerates ROS decomposition. Collectively, empagliflozin treatment contributed to reversal of the circulating progenitor cell imbalance in patients with T2D (Figure 2), correlated with reduced CVD risk as observed in the EMPA-REG OUTCOME trial. Thus, restoration of progenitor cell content in the circulation represented a treatable target to improve vascular homeostasis during T2D.

Reversal of circulating progenitor cell imbalance associated with type 2 diabetes. Circulating progenitor cell imbalance is characterized by increased pro-inflammatory progenitor cells and the depletion of pro-angiogenic haematopoietic and monocytes in the peripheral blood. (A) In individuals with type 2 diabetes, increased circulating ALDHhiSSChi granulocyte precursors and increased ALDHhiSSCmid monocytes with M1 polarization contribute to elevated systemic inflammation associated with endothelial dysfunction. Reduced circulating ALDHhiSSClow cells with pro-angiogenic secretory capacity will interfere with the repair of damaged vessels. After administration of the SGLT2i empagliflozin, decreased circulating ALDHhiSSChi granulocyte content alongside improved ALDHhiSSCmid monocyte balance will reduce systemic inflammation. Increased circulating ALDHhiSSClow cells with CD34/CD133 co-expression will promote vessel repair.
Figure 2

Reversal of circulating progenitor cell imbalance associated with type 2 diabetes. Circulating progenitor cell imbalance is characterized by increased pro-inflammatory progenitor cells and the depletion of pro-angiogenic haematopoietic and monocytes in the peripheral blood. (A) In individuals with type 2 diabetes, increased circulating ALDHhiSSChi granulocyte precursors and increased ALDHhiSSCmid monocytes with M1 polarization contribute to elevated systemic inflammation associated with endothelial dysfunction. Reduced circulating ALDHhiSSClow cells with pro-angiogenic secretory capacity will interfere with the repair of damaged vessels. After administration of the SGLT2i empagliflozin, decreased circulating ALDHhiSSChi granulocyte content alongside improved ALDHhiSSCmid monocyte balance will reduce systemic inflammation. Increased circulating ALDHhiSSClow cells with CD34/CD133 co-expression will promote vessel repair.

In contrast, Bonora et al. reported that in patients with T2D, circulating CD34+ and CD34+/VEGFR2+ cells was not reduced in subjects that received dapagliflozin compared to placebo,67 suggesting the significant reversal of the progenitor cell imbalance observed in our studies66 may not be conserved between different formulations within the SGLT2i class, and/or additional assessment identifying progenitor cell subsets with high ALDH activity expression was required to document circulating progenitor cell recovery.

Gastric bypass surgery improved the circulating progenitor cell imbalance associated with severe obesity

Abnormal adiposity increases the risk of developing T2D and adverse cardiovascular events.68 Roux-en-Y gastric bypass surgery is an effective intervention to induce sustained weight loss compared to non-surgical diet restriction with best medical therapy.69 Remarkably, gastric bypass surgery has also been associated with spontaneous remission of T2D and profound CVD risk reductions.69  ,  70 Because the metabolic and cellular mechanisms driving these benefits were undetermined, we analysed circulating ALDHhi progenitor cell frequencies in normal-weight controls (BMI <25 kg/m2) compared to patients living with severe obesity (BMI >40 kg/m2) prior to and 3 months after gastric bypass surgery.71 Blood from individuals with severe obesity consistently demonstrated elevated circulating ALDHhiSSChi granulocyte precursor content compared to normal-weight controls. Remarkably, normal granulocyte distribution was restored 3 months post-surgery. ALDHhiSSCmid monocytes with pro-inflammatory M1 polarization (CD14+/CD80+ cells) were also elevated in bariatric surgery patients compared to normal-weight controls, and were significantly reduced 3 months after surgery. In contrast, ALDHhiSSCmid monocytes with M2 polarization (CD14+/CD163+) were increased post-surgery. Finally, ALDHhiSSClow cells co-expressing CD34 and CD133 were depleted in individuals with obesity and significantly increased after surgery (Figure 2). Thus, increased inflammatory cell burden and vessel regenerative progenitor subset depletion during obesity was remarkably improved within 3 months of bariatric surgery.

Proposed mechanisms implicated in blood vessel regeneration

While detailed functional evaluation of purified ALDHhi progenitor cell subsets in patients with T2D and obesity remain ongoing in our laboratory, seminal contributions by several groups have investigated further into analogous cellular subtypes integral to endogenous blood vessel repair during T2D. In pre-clinical mouse studies, the Fadini group has clearly established that during the metabolic consequences of T2D, the BM undergoes extensive remodelling, including microvessel rarefaction and fat accumulation, generating a hostile environment for resident progenitor cell differentiation.72 Fadini further suggested that HPC mobilopathy and enhanced myelopoietic differentiation into progeny secreting pro-inflammatory and anti-angiogenic mediators occurred as a result of T2D.73  ,  74 The observed mobilopathy effect was attributed to an accumulation of CD169+ macrophages in the BM that prevented the mobilization of haematopoietic cells.75

Cappellari et al.76 focused on the role of a novel granulocyte subset expressing the integrin receptor CD49d as a potent mediator of vessel repair in response to endothelial damage. CD49d+ granulocytes augmented tubule formation via physical interactions with endothelial cells in vitro, and granulocytes from subjects with T2D had profoundly impaired capacity to stimulate endothelial cell tube formation compared to non-diabetic control populations. CD49d+ granulocytes were 30–40% reduced in the peripheral blood of human subjects with T2D and were two-fold increased after dapagliflozin treatment. This study first established reduced vessel-regenerative granulocyte content during T2D could be modulated by dapagliflozin treatment.

Currently, there is little evidence in the literature to support that SGLT2i or gastric bypass surgery demonstrate direct effects on BM-derived progenitor cells. However, in recent mechanistic studies performed in streptozotocin (STZ)-treated hyperglycaemic mice, Albiero et al.77 demonstrated that despite low levels of SGLT2 receptor expression on BM-derived cells, SGLT2 inhibition with dapagliflozin improved endothelial repair through enhanced mobilization and recruitment of haematopoietic cells from the BM directly to the site of carotid artery injury. In hyperglycaemic mice, dapagliflozin administration reduced the frequency of CD169+ macrophages in mouse BM by 70% and partially rescued the mobilization defect in response to granulocyte colony-stimulating factor. Next, the authors examined the effect of improved haematopoietic cell mobilization after the induction of carotid artery wound healing. Remarkably, re-endothelialization after denudation injury was 10-fold decreased in hyperglycaemic mice, and endothelial healing was significantly improved after dapagliflozin treatment. In elegant experiments using chimeric mice reconstituted with GFP+ BM cells to effectively trace GFP+ cells localized to the site of injury, reduced recruitment of GFP+ cells to the site of carotid injury observed in STZ-treated diabetic chimeras was reversed by dapagliflozin treatment. Mature GFP+ monocytes and neutrophils were abundant in the damaged carotid artery, with both F4/80+ macrophage and CD49d+ granulocyte recruitment significantly increased by dapagliflozin treatment. Thus, monocyte and CD49d+ granulocyte mobilization, recruitment, and adhesion at the site of injury promoted re-endothelialization after dapagliflozin administration, identifying CD49d+ granulocytes as a novel therapeutic target to promote endothelial repair during diabetes. These findings suggested that modulation of granulocyte subset generation and mobilization using SGLT2i treatment may represent a therapeutic strategy to repair damaged vessels during T2D.

Clinical perspectives and future directions

Novel pharmacological interventions to reverse the circulating progenitor cell imbalance during T2D and obesity

Rational selection of therapeutic agents with the potential to improve endogenous vessel repair has been repeatedly demonstrated by agents that promote glucose control. However, in our relatively small substudy of 30 patients, ongoing metformin or insulin therapy did not improve the circulating progenitor cell imbalance phenotype observed during T2D,59 despite previous reports that these agents improve HSPC content in the peripheral blood.28 In addition, pioglitazone and dipeptidyl peptidase-4 inhibitors have been previously reported to directly improve CD34+ cell trafficking.78  ,  79 The widespread metabolic stabilization following gastric bypass surgery has also provided insight into potential mechanisms driving improved circulating progenitor cell content. Even before significant weight loss has occurred after surgery, ∼40% patients experienced metabolic stabilization including improved glycaemic control and increased insulin sensitivity that can result in discontinuation of T2D medication.69 Due to direct gastric emptying into a distal segment of the gastrointestinal tract, gastric bypass surgery consistently results in a sharp postprandial rise in gut peptides, including the incretin hormone and satiety protein glucagon-like peptide-1 (GLP-1). A recent meta-analysis evaluating 368 individuals across 24 studies after gastric bypass surgery indicated that postprandial GLP-1 levels were significantly increased following surgery, providing evidence that GLP-1 receptor (GLP-1R) signalling axis may play an important role in the metabolic benefits observed following bariatric surgery.80 GLP-1 is a hormone produced primarily in the ileum and colon and secreted into the circulation in response to increased glucose load. GLP-1 enhances insulin secretion by activating GLP-1R on the surface of β cells in a glucose-dependent manner. GLP-1R is expressed throughout the gastrointestinal tract and in the satiety control centre of the hypothalamus. Therefore, in addition to insulinotropic effects, GLP-1R activity has been shown to delay gastric emptying, increase appetite suppression, and reduce glucagon activity in the clinical setting. Furthermore, GLP-1R is surprisingly expressed on many mesodermal cell types, including arterial smooth muscle cells, endothelial cells, and various haematopoietic cell lineages.81 In addition, animal models have shown infusion of GLP-1 increased HPC proliferation and differentiation,82  ,  83 protected of endothelial cells from oxidative stress,84 and improved endothelial cell function.85 We postulate that increased GLP-1 release following gastric bypass surgery may contribute towards improved vessel repair via modulation of the progenitor cell imbalance paradigm86.

Over the past 12 years, recombinant human GLP-1R agonists including liraglutide (2009), dulaglutide (2014), and semaglutide (2017), among others, have been sequentially released. Delivery of GLP-1R agonists has been by weekly subcutaneous injections. However, novel advancements in the formulation of oral semaglutide (2019) have recently been developed.87 Notably, the LEADER trial revealed that the GLP-1R agonist (liraglutide) significantly reduced MACE and all-cause mortality in individuals with T2D.88 Because gastric bypass surgery is highly invasive and recommended only to patients meeting the selection criteria for bariatric surgery (typically BMI ≥40 kg/m2), and because liraglutide is indicated for weight loss during obesity (BMI >30 kg/m2), we have initiated a clinical trial to determine whether treatment with liraglutide can improve circulating progenitor cell content in patients with more moderate obesity (BMI 30–35 kg/m2).

Extracellular vesicles: a novel therapeutic opportunity to induce vessel repair

Human HPC, EPC, and MSC are highly secretory and abundantly produce extracellular vesicles (EVs), including exosomes and microvesicles, that contain potent regenerative cargo in the form of proteins, lipids, and microRNAs. EVs are released by membrane blebbing or exocytotic mechanisms and used for intracellular communication and delivery of signals over long distances via the blood stream.89 A primary target for microparticle uptake is the vascular endothelial lining. As such, investigating the role of extracellular vesicles in vessel repair is warranted as an exciting modality to improve blood vessel regeneration during chronic T2D and obesity.

EVs secreted by CD34+ HPC have been evaluated for their ability to induce blood vessel growth.89–91 Furthermore, the pro-vascular functions of a plethora of miRNAs have been associated with endothelial cell homeostasis, including repression of senescence, increasing adhesion molecule expression, and activation of the developmental Wnt-signalling pathway central to progenitor cell differentiation. Specifically, van Balkom et al.91 demonstrated that miR-214 plays a central role in the induction of angiogenesis in mice. Because CD34+ HPC stimulate capillary formation through pro-angiogenic cytokine release, Mathiyalagan et al.90 recently examined the exosomal contents of CD34+ cells in mice after femoral artery ligation-induced unilateral hind-limb ischaemia. Excitingly, mice injected with purified CD34+ cell exosomes demonstrated improved hind-limb perfusion and preserved muscle mass. MicroRNA screening uncovered CD34+ cell exosomes contained high levels of miR-10a and miR-130 with previously demonstrated anti-fibrotic activities in the ischaemic myocardium.90  ,  92  ,  93 Fibrotic remodelling after myocardial infarct can limit stroke volume and cardiac output due to myocardial thickening and progression towards heart failure.94  ,  95 Furthermore, fibrosis in the vasculature can lead to hypertension and increased mechanical stress on the heart.94  ,  95 Although the use of designer EVs to impact blood vessel regeneration in the clinical setting requires further mechanistic experimentation and extensive regulatory oversight, the potential to improve vessel health and augment cardiac output after myocardial infarction by the delivery of progenitor cell-engineered EVs represents an exciting opportunity to combat CVD in the future.

Supplementation of vascular grafts with regenerative progenitor cells

Management of cardiovascular remodelling after sub-lethal myocardial infarction remains a critical challenge worldwide. Native vessel grafts from autologous mammary arteries or saphenous veins during coronary artery bypass graft surgery are often unsuitable for grafting, fuelling the search for a more mechanically robust alternative.96  ,  97 Dahl et al.98  ,  99 elegantly reviewed the advancements made in the previous three decades in the generation of bioengineered vascular grafts. Most processes involve the creation of new vascular conduits employ in vitro growth from autologous progenitor cells grown in 3D bioscaffolds. Hoerstrup et al.100 demonstrated 100% primary patency at 20–100 weeks following pulmonary artery replacement with bioengineered vascular grafts in bovine models. While the generation of autologous grafts using progenitor cells in vitro has the potential to confer benefit in the maintenance and integration of vascular grafts, there have been challenges with the time needed to generate the vessels. For example, the use of bioengineered vascular grafts could increase patient wait time by weeks or months to grow a sufficient conduit, calling for the examination of more efficient vessel growth strategies in vitro. As such, the applicability of engineered vessels as an alternative to native vessels during revascularization procedures remains arguably premature.

A more immediate strategy to improve the integrity of vessels appropriate for grafting may involve supplementation of compromised grafts with autologous ALDHhi subpopulations, CD49d+ granulocytes, M2-polarized monocytes, to promote angiogenic or arteriogenic remodelling within the graft itself. Consequently, improving upon transplanted vessel survival and the risk of ischaemia re-establishment, specifically in micro-vascular capillaries, may be instrumental in improving vessel patency in the future. Finally, supplementation of native vessels with bioengineered materials such as autologous pro-angiogenic progenitor cell subsets or designer EVs could serve to improve graft integrity while stimulating the repair of native vessels. This union could bolster graft stability to become more effective for functional revascularization after surgery.

Concluding remarks

Pro-vascular progenitor cells transplantation has been proposed as a novel modality to treat CVD through the induction vessel regeneration. Unfortunately, hyperglycaemia and excess adiposity associated with T2D and obesity generate a damaging microenvironment in the BM and impact the production and mobilization of progenitor cells required to combat vessel damage (See Graphical Abstract). Assessment of ALDHhi progenitor cell subsets in the circulation of patients with T2D and obesity establish that pro-oxidative and pro-inflammatory microenvironment negatively impact circulating pro-angiogenic progenitor cell content. Early intervention with strategies that prevent or reverse abnormalities in the circulating progenitor cell pool represents an under recognized mechanism to effectively augment vessel repair and regeneration. Therefore, early intervention in patients with developing T2D and obesity may provide immediately translatable therapeutic options to reduce blood vessel damage and preserve blood vessel health in patients with T2D or obesity.

Acknowledgements

The authors gratefully acknowledge Gail Rudkevich in the generation of the figures presented.

Conflict of interest: D.A.H. does not have any conflict of interest. S.V. holds a Tier 1 Canada Research Chair in Cardiovascular Surgery and reports receiving research grants and/or speaking honoraria from Amarin, Amgen, AstraZeneca, Bayer, Boehringer Ingelheim, Bristol-Myers Squibb, Eli Lilly, EOCI Pharmacomm Ltd, HLS Therapeutics, Janssen, Merck, Novartis, Novo Nordisk, Pfizer, PhaseBio, Sanofi, Sun Pharmaceuticals, and the Toronto Knowledge Translation Working Group. D.B. discloses the following relationships—Advisory Board: Cardax, CellProthera, Cereno Scientific, Elsevier Practice Update Cardiology, Janssen, Level Ex, Medscape Cardiology, MyoKardia, Novo Nordisk, PhaseBio, PLx Pharma, and Regado Biosciences; Board of Directors: Boston VA Research Institute, Society of Cardiovascular Patient Care, TobeSoft; Chair: American Heart Association Quality Oversight Committee; Data Monitoring Committees: Baim Institute for Clinical Research (formerly Harvard Clinical Research Institute, for the PORTICO trial, funded by St. Jude Medical, now Abbott), Cleveland Clinic (including for the ExCEED trial, funded by Edwards), Contego Medical (Chair, PERFORMANCE 2), Duke Clinical Research Institute, Mayo Clinic, Mount Sinai School of Medicine (for the ENVISAGE trial, funded by Daiichi Sankyo), Population Health Research Institute; Honoraria: American College of Cardiology (Senior Associate Editor, Clinical Trials and News, ACC.org; Vice-Chair, ACC Accreditation Committee), Baim Institute for Clinical Research (formerly Harvard Clinical Research Institute; RE-DUAL PCI clinical trial steering committee funded by Boehringer Ingelheim; AEGIS-II executive committee funded by CSL Behring), Belvoir Publications (Editor in Chief, Harvard Heart Letter), Canadian Medical and Surgical Knowledge Translation Research Group (clinical trial steering committees), Duke Clinical Research Institute (clinical trial steering committees, including for the PRONOUNCE trial, funded by Ferring Pharmaceuticals), HMP Global (Editor-in-Chief, Journal of Invasive Cardiology), Journal of the American College of Cardiology (Guest Editor; Associate Editor), K2P (Co-Chair, interdisciplinary curriculum), Level Ex, Medtelligence/ReachMD (CME steering committees), MJH Life Sciences, Population Health Research Institute (for the COMPASS operations committee, publications committee, steering committee, and USA national co-leader, funded by Bayer), Slack Publications (Chief Medical Editor, Cardiology Today’s Intervention), Society of Cardiovascular Patient Care (Secretary/Treasurer), WebMD (CME steering committees); Other: Clinical Cardiology (Deputy Editor), NCDR-ACTION Registry Steering Committee (Chair), VA CART Research and Publications Committee (Chair); Research Funding: Abbott, Afimmune, Amarin, Amgen, AstraZeneca, Bayer, Boehringer Ingelheim, Bristol-Myers Squibb, Cardax, CellProthera, Cereno Scientific, Chiesi, CSL Behring, Eisai, Ethicon, Ferring Pharmaceuticals, Forest Laboratories, Fractyl, Garmin, HLS Therapeutics, Idorsia, Ironwood, Ischemix, Janssen, Lexicon, Lilly, Medtronic, MyoKardia, Novartis, Novo Nordisk, Owkin, Pfizer, PhaseBio, PLx Pharma, Regeneron, Roche, Sanofi, Synaptic, The Medicines Company, 89Bio; Royalties: Elsevier (Editor, Cardiovascular Intervention: A Companion to Braunwald’s Heart Disease); Site Co-Investigator: Abbott, Biotronik, Boston Scientific, CSI, St. Jude Medical (now Abbott), Svelte; Trustee: American College of Cardiology; and Unfunded Research: FlowCo, Merck, Takeda. E.B. and D.C.T. do not report potential conflicts of interest relevant to this article. F.C. reports receiving research grants from Swedish Research Council, Swedish Heart & Lung Foundation, and King Gustav V and Queen Victoria Foundation, as well as speaking/advisory board honoraria from Abbott, AstraZeneca, Bayer, Bristol-Myers Squibb, Merck Sharp & Dohme, Mundipharma, Novo Nordisk, Boehringer Ingelheim, and Pfizer.

Data availability

The data in this article will be shared upon reasonable request to the corresponding author.

References

1

Kaptoge
 
S
,
Pennells
 
L
,
De Bacquer
 
D
 et al.  
World Health Organization cardiovascular disease risk charts: revised models to estimate risk in 21 global regions
.
Lancet Glob Health
 
2019
;
7
:
e1332
e1345
.

2

Herrington
 
W
,
Lacey
 
B
,
Sherliker
 
P
,
Armitage
 
J
,
Lewington
 
S.
 
Epidemiology of atherosclerosis and the potential to reduce the global burden of atherothrombotic disease
.
Circ Res
 
2016
;
118
:
535
546
.

3

Roth
 
GA
,
Abate
 
D
,
Abate
 
KH
 et al.  
Global, regional, and national age-sex-specific mortality for 282 causes of death in 195 countries and territories, 1980–2017: a systematic analysis for the Global Burden of Disease Study 2017
.
Lancet
 
2018
;
392
:
1736
1788
.

4

Townsend
 
N
,
Nichols
 
M
,
Scarborough
 
P
,
Rayner
 
M.
 
Cardiovascular disease in Europe - Epidemiological update 2015
.
Eur Heart J
 
2015
;
36
:
2696
2705
.

5

Poirier
 
P
,
Giles
 
TD
,
Bray
 
GA
 et al. ;
Obesity Committee of the Council on Nutrition, Physical Activity, and Metabolism
.
Obesity and cardiovascular disease: pathophysiology, evaluation, and effect of weight loss: an update of the 1997 American Heart Association Scientific Statement on obesity and heart disease from the Obesity Committee of the Council on Nutrition, Physical Activity, and Metabolism
.
Circulation
 
2006
;
113
:
898
918
.

6

Bommer
 
C
,
Heesemann
 
E
,
Sagalova
 
V
 et al.  
The global economic burden of diabetes in adults aged 20–79 years: a cost-of-illness study
.
Lancet Diabetes Endocrinol
 
2017
;
5
:
423
430
.

7

Hex
 
N
,
Bartlett
 
C
,
Wright
 
D
,
Taylor
 
M
,
Varley
 
D.
 
Estimating the current and future costs of Type1 and Type2 diabetes in the UK, including direct health costs and indirect societal and productivity costs
.
Diabet Med
 
2012
;
29
:
855
862
.

8

Mechanick
 
JI
,
Farkouh
 
ME
,
Newman
 
JD
,
Garvey
 
WT.
 
Cardiometabolic-based chronic disease, adiposity and dysglycemia drivers: JACC state-of-the-art review
.
J Am Coll Cardiol
 
2020
;
75
:
525
538
.

9

Paneni
 
F
,
Beckman
 
JA
,
Creager
 
MA
,
Cosentino
 
F.
 
Diabetes and vascular disease: pathophysiology, clinical consequences, and medical therapy: part I
.
Eur Heart J
 
2013
;
34
:
2436
2446
.

10

Costantino
 
S
,
Paneni
 
F
,
Virdis
 
A
 et al.  
Interplay among H3K9-editing enzymes SUV39H1, JMJD2C and SRC-1 drives p66Shc transcription and vascular oxidative stress in obesity
.
Eur Heart J
 
2019
;
40
:
383
391
.

11

Costantino
 
S
,
Paneni
 
F
,
Battista
 
R
 et al.  
Impact of glycemic variability on chromatin remodeling, oxidative stress, and endothelial dysfunction in patients with type 2 diabetes and with target HbA1c levels
.
Diabetes
 
2017
;
66
:
2472
2482
.

12

Hussain
 
S
,
Khan
 
AW
,
Akhmedov
 
A
 et al.  
Hyperglycemia induces myocardial dysfunction via epigenetic regulation of JunD
.
Circ Res
 
2020
;
127
:
1261
1273
.

13

Paneni
 
F
,
Costantino
 
S
,
Castello
 
L
 et al.  
Targeting prolyl-isomerase Pin1 prevents mitochondrial oxidative stress and vascular dysfunction: insights in patients with diabetes
.
Eur Heart J
 
2015
;
36
:
817
828
.

14

Krams
 
R
,
Back
 
M.
 
ESC Textbook of Vascular Biology
.
Oxford
:
Oxford University Press (OUP)
;
2017
.

15

Terenzi
 
DC
,
Trac
 
JZ
,
Teoh
 
H
 et al.  
Vascular regenerative cell exhaustion in diabetes: translational opportunities to mitigate cardiometabolic risk
.
Trends Mol Med
 
2019
;
25
:
640
655
.

16

Asahara
 
T
,
Murohara
 
T
,
Sullivan
 
A
 et al.  
Isolation of putative progenitor endothelial cells for angiogenesis
.
Science
 
1997
;
275
:
964
967
.

17

Isner
 
JM
,
Asahara
 
T.
 
Angiogenesis and vasculogenesis as therapeutic strategies for postnatal neovascularization
.
J Clin Invest
 
1999
;
103
:
1231
1236
.

18

Murray
 
PJ
,
Allen
 
JE
,
Biswas
 
SK
,
Fisher
 
EA
 et al.  
Macrophage activation and polarization: nomenclature and experimental guidelines
.
Immunity
 
2014
;
41
:
14
20
.

19

Mosser
 
DM
,
Edwards
 
JP.
 
Exploring the full spectrum of macrophage activation
.
Nat Rev Immunol
 
2008
;
8
:
958
969
.

20

Ruiz De Almodovar
 
C
,
Luttun
 
A
,
Carmeliet
 
P.
 
An SDF-1 trap for myeloid cells stimulates angiogenesis
.
Cell
 
2006
;
124
:
18
21
.

21

Asahara
 
T
,
Masuda
 
H
,
Takahashi
 
T
 et al.  
Bone marrow origin of endothelial progenitor cells responsible for postnatal vasculogenesis in physiological and pathological neovascularization
.
Circ Res
 
1999
Aug 6;
85
:
221
228
.

22

Vasa
 
M
,
Fichtlscherer
 
S
,
Aicher
 
A
 et al.  
Number and migratory activity of circulating endothelial progenitor cells inversely correlate with risk factors for coronary artery disease
.
Circ Res
 
2001
;
89
:
E1
E7
.

23

Werner
 
N
,
Kosiol
 
S
,
Schiegl
 
T
 et al.  
Circulating endothelial progenitor cells and cardiovascular outcomes
.
N Engl J Med
 
2005
;
353
:
999
1007
.

24

Zafar
 
N
,
Krishnasamy
 
SS
,
Shah
 
J
,
Rai
 
SN
 et al.  
Circulating angiogenic stem cells in type 2 diabetes are associated with glycemic control and endothelial dysfunction
.
PLoS One
 
2018
;
13
:
e0205851
.

25

Fadini
 
GP
,
Miorin
 
M
,
Facco
 
M
 et al.  
Circulating endothelial progenitor cells are reduced in peripheral vascular complications of type 2 diabetes mellitus
.
J Am Coll Cardiol
 
2005
May 3;
45
:
1449
1457
.

26

Fadini
 
G
,
de Kreutzenberg
 
S
,
Coracina
 
A
 et al.  
Circulating CD34+ cells, metabolic syndrome, and cardiovascular risk
.
Eur Heart J
 
2006
;
27
:
2247
2255
.

27

Fadini
 
GP
,
Bonora
 
BM
,
Marcuzzo
 
G
 et al.  
Circulating stem cells associate with adiposity and future metabolic deterioration in healthy subjects
.
J Clin Endocrinol Metab
 
2015
;
100
:
4570
4578
.

28

Fadini
 
GP
,
Mehta
 
A
,
Dhindsa
 
DS
 et al.  
Circulating stem cells and cardiovascular outcomes: from basic science to the clinic
.
Eur Heart J
 
2020
;
41
:
4271
4282
.

29

Passegué
 
E
,
Wagers
 
AJ
,
Giuriato
 
S
,
Anderson
 
WC
,
Weissman
 
IL.
 
Global analysis of proliferation and cell cycle gene expression in the regulation of hematopoietic stem and progenitor cell fates
.
J Exp Med
 
2005
;
202
:
1599
1611
.

30

Takakura
 
N
,
Watanabe
 
T
,
Suenobu
 
S
 et al.  
A role for hematopoietic stem cells in promoting angiogenesis
.
Cell
 
2000
;
102
:
199
209
.

31

Urbich
 
C
,
Heeschen
 
C
,
Aicher
 
A
,
Dernbach
 
E
,
Zeiher
 
AM
,
Dimmeler
 
S.
 
Relevance of monocytic features for neovascularization capacity of circulating endothelial progenitor cells
.
Circulation
 
2003
;
108
:
2511
2516
.

32

Yoder
 
MC
,
Mead
 
LE
,
Prater
 
D
 et al.  
Redefining endothelial progenitor cells via clonal analysis and hematopoietic stem/progenitor cell principals
.
Blood
 
2007
;
109
:
1801
1809
.

33

Deindl
 
E
,
Buschmann
 
I
,
Hoefer
 
IE
 et al.  
Role of ischemia and of hypoxia-inducible genes in arteriogenesis after femoral artery occlusion in the rabbit
.
Circ Res
 
2001
;
89
:
779
786
.

34

Awad
 
O
,
Dedkov
 
EI
,
Jiao
 
C
,
Bloomer
 
S
,
Tomanek
 
RJ
,
Schatteman
 
GC.
 
Differential healing activities of CD34+ and CD14+ endothelial cell progenitors
.
Arterioscler Thromb Vasc Biol
 
2006
;
26
:
758
764
.

35

Michaud
 
SE
,
Ménard
 
C
,
Guy
 
LG
,
Gennaro
 
G
,
Rivard
 
A.
 
Inhibition of hypoxia-induced angiogenesis by cigarette smoke exposure: impairment of the HIF-1alpha/VEGF pathway
.
FASEB J
 
2003
;
17
:
1150
1152
.

36

Schiekofer
 
S
,
Galasso
 
G
,
Sato
 
K
,
Kraus
 
BJ
,
Walsh
 
K.
 
Impaired revascularization in a mouse model of type 2 diabetes is associated with dysregulation of a complex angiogenic-regulatory network
.
Arterioscler Thromb Vasc Biol
 
2005
;
25
:
1603
1609
.

37

Van Golde
 
JM
,
Ruiter
 
MS
,
Schaper
 
NC
 et al.  
Impaired collateral recruitment and outward remodeling in experimental diabetes
.
Diabetes
 
2008
;
57
:
2818
2823
.

38

Yan
 
J
,
Tie
 
G
,
Park
 
B
,
Yan
 
Y
,
Nowicki
 
PT
,
Messina
 
LM.
 
Recovery from hind limb ischemia is less effective in type 2 than in type 1 diabetic mice: roles of endothelial nitric oxide synthase and endothelial progenitor cells
.
J Vasc Surg
 
2009
;
50
:
1412
1422
.

39

Murray
 
PJ.
 
Macrophage Polarization
.
Annu Rev Physiol
 
2017
;
79
:
541
566
.

40

Troidl
 
C
,
Jung
 
G
,
Troidl
 
K
 et al.  
The temporal and spatial distribution of macrophage subpopulations during arteriogenesis
.
Curr Vasc Pharmacol
 
2012
;
11
:
5
12
.

41

Takeda
 
Y
,
Costa
 
S
,
Delamarre
 
E
 et al.  
Macrophage skewing by Phd2 haplodeficiency prevents ischaemia by inducing arteriogenesis
.
Nature
 
2011
;
479
:
122
126
.

42

Sherman
 
SE
,
Kuljanin
 
M
,
Cooper
 
TT
,
Putman
 
DM
,
Lajoie
 
GA
,
Hess
 
DA.
 
High aldehyde dehydrogenase activity identifies a subset of human mesenchymal stromal cells with vascular regenerative potential
.
Stem Cells
 
2017
;
35
:
1542
1553
.

43

Putman
 
DM
,
Cooper
 
TT
,
Sherman
 
SE
 et al.  
Expansion of umbilical cord blood aldehyde dehydrogenase expressing cells generates myeloid progenitor cells that stimulate limb revascularization
.
Stem Cells Transl Med
 
2017
;
6
:
1607
1619
.

44

Putman
 
DM
,
Liu
 
KY
,
Broughton
 
HC
,
Bell
 
GI
,
Hess
 
DA.
 
Umbilical cord blood-derived aldehyde dehydrogenase-expressing progenitor cells promote recovery from acute ischemic injury
.
Stem Cells
 
2012
;
30
:
2248
2260
.

45

Storms
 
RW
,
Trujillo
 
AP
,
Springer
 
JB
 et al.  
Isolation of primitive human hematopoietic progenitors on the basis of aldehyde dehydrogenase activity
.
Proc Natl Acad Sci USA
 
1999
;
96
:
9118
9123
.

46

Storms
 
RW
,
Green
 
PD
,
Safford
 
KM
 et al.  
Distinct hematopoietic progenitor compartments are delineated by the expression of aldehyde dehydrogenase and CD34
.
Blood
 
2005
;
106
:
95
102
.

47

Hess
 
DA
,
Meyerrose
 
TE
,
Wirthlin
 
L
 et al.  
Functional characterization of highly purified human hematopoietic repopulating cells isolated according to aldehyde dehydrogenase activity
.
Blood
 
2004
;
104
:
1648
1655
.

48

Hess
 
DA
,
Wirthlin
 
L
,
Craft
 
TP
 et al.  
Selection based on CD133 and high aldehyde dehydrogenase activity isolates long-term reconstituting human hematopoietic stem cells
.
Blood
 
2006
;
107
:
2162
2169
.

49

Capoccia
 
BJ
,
Robson
 
DL
,
Levac
 
KD
 et al.  
Revascularization of ischemic limbs after transplantation of human bone marrow cells with high aldehyde dehydrogenase activity
.
Blood
 
2009
;
113
:
5340
5351
.

50

Perin
 
EC
,
Silva
 
G
,
Gahremanpour
 
A
 et al.  
A randomized, controlled study of autologous therapy with bone marrow-derived aldehyde dehydrogenase bright cells in patients with critical limb ischemia
.
Catheter Cardiovasc Interv
 
2011
;
78
:
1060
1067
.

51

Perin
 
EC
,
Murphy
 
MP
,
March
 
KL
 et al. ;
Cardiovascular Cell Therapy Research Network (CCTRN)
.
Evaluation of cell therapy on exercise performance and limb perfusion in peripheral artery disease: the CCTRN PACE Trial (Patients with Intermittent Claudication Injected with ALDH Bright Cells)
.
Circulation
 
2017
;
135
:
1417
1428
.

52

Putman
 
DM
,
Cooper
 
TT
,
Sherman
 
SE
 et al.  
Expansion of umbilical cord blood aldehyde dehydrogenase expressing cells generates myeloid progenitor cells that stimulate limb revascularization
.
Stem Cells Transl Med
 
2017
;
6
:
1607
1619
.

53

Nakamura-Ishizu
 
A
,
Takizawa
 
H
,
Suda
 
T.
 
The analysis, roles and regulation of quiescence in hematopoietic stem cells
.
Development
 
2014
;
141
:
4656
4666
.

54

Fadini
 
GP
,
Sartore
 
S
,
Schiavon
 
M
 et al.  
Diabetes impairs progenitor cell mobilisation after hindlimb ischaemia-reperfusion injury in rats
.
Diabetologia
 
2006
;
49
:
3075
3084
.

55

Fadini
 
GP
,
Sartore
 
S
,
Albiero
 
M
 et al.  
Number and function of endothelial progenitor cells as a marker of severity for diabetic vasculopathy
.
Arterioscler Thromb Vasc Biol
 
2006
;
26
:
2140
2146
.

56

Hess
 
DA
,
Hegele
 
RA.
 
Linking diabetes with oxidative stress, adipokines, and impaired endothelial precursor cell function
.
Can J Cardiol
 
2012
;
28
:
629
630
.

57

Jialal
 
I
,
Devaraj
 
S
,
Singh
 
U
,
Huet
 
BA.
 
Decreased number and impaired functionality of endothelial progenitor cells in subjects with metabolic syndrome: implications for increased cardiovascular risk
.
Atherosclerosis
 
2010
;
211
:
297
302
.

58

Qadura
 
M
,
Terenzi
 
DC
,
Verma
 
S
,
Al-Omran
 
M
,
Hess
 
DA.
 
Concise review: cell therapy for critical limb ischemia: an integrated review of preclinical and clinical studies
.
Stem Cells
 
2018
;
36
:
161
171
.

59

Terenzi
 
DC
,
Al-Omran
 
M
,
Quan
 
A
,
Teoh
 
H
,
Verma
 
S
,
Hess
 
DA.
 
Circulating pro-vascular progenitor cell depletion during type 2 diabetes
.
JACC Basic Transl Sci
 
2019
;
4
:
98
112
.

60

Nagareddy
 
PR
,
Sreejit
 
G
,
Lee
 
MK
 et al.  
Inflammasome-primed neutrophils return to the bone marrow to stimulate leukocytosis following myocardial infarction
.
Circ Res
 
2020
;
127
:
A228
.

61

Hoyer
 
FF
,
Zhang
 
X
,
Coppin
 
E
 et al.  
Bone marrow endothelial cells regulate myelopoiesis in diabetes mellitus
.
Circulation
 
2020
;
142
:
244
258
.

62

Nagareddy
 
PR
,
Kraakman
 
M
,
Masters
 
SL
 et al.  
Adipose tissue macrophages promote myelopoiesis and monocytosis in obesity
.
Cell Metab
 
2014
;
19
:
821
835
.

63

Ferrannini
 
E
,
Solini
 
A.
 
SGLT2 inhibition in diabetes mellitus: rationale and clinical prospects
.
Nat Rev Endocrinol
 
2012
;
8
:
495
502
.

64

Zinman
 
B
,
Wanner
 
C
,
Lachin
 
JM
 et al. ;
EMPA-REG OUTCOME Investigators
.
Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes
.
N Engl J Med
 
2015
;
373
:
2117
2128
.

65

Keats
 
EC
,
Dominguez
 
JM
,
Grant
 
MB
,
Khan
 
ZA.
 
Switch from canonical to noncanonical Wnt signaling mediates high glucose-induced adipogenesis
.
Stem Cells
 
2014
;
32
:
1649
1660
.

66

Hess
 
DA
,
Terenzi
 
DC
,
Trac
 
JZ
,
Quan
 
A
 et al.  
SGLT2 inhibition with empagliflozin increases circulating provascular progenitor cells in people with type 2 diabetes mellitus
.
Cell Metab
 
2019
;
30
:
609
613
.

67

Bonora
 
BM
,
Cappellari
 
R
,
Albiero
 
M
,
Avogaro
 
A
,
Fadini
 
GP.
 
Effects of SGLT2 inhibitors on circulating stem and progenitor cells in patients with type 2 diabetes
.
J Clin Endocrinol Metab
 
2018
;
103
:
3773
3782
.

68

Mechanick
 
JI
,
Farkouh
 
ME
,
Newman
 
JD
,
Garvey
 
WT.
 
Cardiometabolic-based chronic disease, adiposity and dysglycemia drivers: JACC state-of-the-art review
.
J Am Coll Cardiol
 
2020
;
75
:
525
538
.

69

Schauer
 
PR
,
Bhatt
 
DL
,
Kirwan
 
JP
 et al.  
Bariatric surgery versus intensive medical therapy for diabetes — 5-year outcomes
.
N Engl J Med
 
2017
;
376
:
641
651
.

70

Adams
 
TD
,
Arterburn
 
DE
,
Nathan
 
DM
,
Eckel
 
RH.
 
Clinical outcomes of metabolic surgery: microvascular and macrovascular complications
.
Diabetes Care
 
2016
;
39
:
912
923
.

71

Hess
 
DA
,
Trac
 
JZ
,
Glazer
 
SA
 et al.  
Vascular risk reduction in obesity through reduced granulocyte burden and improved angiogenic monocyte content following bariatric surgery
.
Cell Reports Med
 
2020
;
1
:
100018
.

72

Fadini
 
GP
,
Spinetti
 
G
,
Santopaolo
 
M
,
Madeddu
 
P.
 
Impaired regeneration contributes to poor outcomes in diabetic peripheral artery disease
.
Arterioscler Thromb Vasc Biol
 
2020
;
40
:
34
44
.

73

Fadini
 
GP
,
Albiero
 
M
,
De Kreutzenberg
 
SV
 et al.  
Diabetes impairs stem cell and proangiogenic cell mobilization in humans
.
Diabetes Care
 
2013
;
36
:
943
949
.

74

DiPersio
 
JF.
 
Diabetic stem-cell “mobilopathy”
.
N Engl J Med
 
2011
;
365
:
2536
2538
.

75

Albiero
 
M
,
Ciciliot
 
S
,
Tedesco
 
S
 et al.  
Diabetes-associated myelopoiesis drives stem cell mobilopathy through an OSM-p66Shc signaling pathway
.
Diabetes
 
2019
;
68
:
1303
1314
.

76

Cappellari
 
R
,
D’Anna
 
M
,
Menegazzo
 
L
 et al.  
Diabetes mellitus impairs circulating proangiogenic granulocytes
.
Diabetologia
 
2020
Sep 1;
63
:
1872
1884
.

77

Albiero
 
M
,
Tedesco
 
S
,
Amendolagine
 
FI
 et al.  
Inhibition of SGLT-2 rescues bone marrow cell traffic for vascular repair. Role of glucose control and ketogenesis
.
Diabetes
 
2021
;
70
:
1767
1779
.

78

Fadini
 
GP
,
Albiero
 
M
,
Seeger
 
F
 et al.  
Stem cell compartmentalization in diabetes and high cardiovascular risk reveals the role of DPP-4 in diabetic stem cell mobilopathy
.
Basic Res Cardiol
 
2013
;
108
:
313
.

79

Tedesco
 
S
,
Ciciliot
 
S
,
Menegazzo
 
L
 et al.  
Pharmacologic PPAR-γ activation reprograms bone marrow macrophages and partially rescues HSPC mobilization in human and murine diabetes
.
Diabetes
 
2020
;
69
:
1562
1572
.

80

Jirapinyo
 
P
,
Jin
 
DX
,
Qazi
 
T
,
Mishra
 
N
,
Thompson
 
CC.
 
A meta-analysis of GLP-1 after Roux-En-Y gastric bypass: impact of surgical technique and measurement strategy
.
Obes Surg
 
2018
;
28
:
615
626
.

81

Pyke
 
C
,
Heller
 
RS
,
Kirk
 
RK
 et al.  
GLP-1 receptor localization in monkey and human tissue: novel distribution revealed with extensively validated monoclonal antibody
.
Endocrinology
 
2014
;
155
:
1280
1290
.

82

Xie
 
XY
,
Mo
 
ZH
,
Chen
 
K
,
He
 
HH
,
Xie
 
YH.
 
Glucagon-like peptide-1 improves proliferation and differentiation of endothelial progenitor cells via upregulating VEGF generation
.
Med Sci Monit
 
2011
;
17
:
BR35
BR41
.

83

Zhu
 
E
,
Hu
 
L
,
Wu
 
H
 et al.  
Dipeptidyl peptidase-4 regulates hematopoietic stem cell activation in response to chronic stress
.
J Am Heart Assoc
 
2017
;
6
:
e006394
.

84

Cai
 
X
,
She
 
M
,
Xu
 
M
 et al.  
GLP-1 treatment protects endothelial cells from oxidative stress-induced autophagy and endothelial dysfunction
.
Int J Biol Sci
 
2018
;
14
:
1696
1708
.

85

Helmstädter
 
J
,
Frenis
 
K
,
Filippou
 
K
 et al.  
Endothelial GLP-1 (glucagon-like peptide-1) receptor mediates cardiovascular protection by liraglutide in mice with experimental arterial hypertension
.
Arterioscler Thromb Vasc Biol
 
2020
;
40
:
145
158
.

86

Bakbak
 
E
,
Terenzi
 
DC
,
Trac
 
JZ
 et al.  
Lessons from bariatric surgery: Can increased GLP-1 enhance vascular repair during cardiometabolic-based chronic disease?
.
Rev Endocr Metab Disord
 
2021
; 34228302

87

Husain
 
M
,
Birkenfeld
 
AL
,
Donsmark
 
M
 et al. ;
PIONEER 6 Investigators
.
Oral semaglutide and cardiovascular outcomes in patients with type 2 diabetes
.
N Engl J Med
 
2019
;
381
:
841
851
.

88

Marso
 
SP
,
Daniels
 
GH
,
Brown-Frandsen
 
K
 et al.  
LEADER steering committee; leader trial investigators. liraglutide and cardiovascular outcomes in type 2 diabetes
.
N Engl J Med
 
2016
;
375
:
311
322
.

89

Todorova
 
D
,
Simoncini
 
S
,
Lacroix
 
R
,
Sabatier
 
F
,
Dignat-George
 
F.
 
Extracellular vesicles in angiogenesis
.
Circ Res
 
2017
;
120
:
1658
1673
.

90

Mathiyalagan
 
P
,
Liang
 
Y
,
Kim
 
D
 et al.  
Angiogenic mechanisms of human CD34+stem cell exosomes in the repair of ischemic hindlimb
.
Circ Res
 
2017
;
120
:
1466
1476
.

91

van Balkom
 
BWM
,
de Jong
 
OG
,
Smits
 
M
 et al.  
Endothelial cells require miR-214 to secrete exosomes that suppress senescence and induce angiogenesis in human and mouse endothelial cells
.
Blood
 
2013
;
121
:
3997
4006
.

92

Dong
 
J
,
Zhang
 
Z
,
Huang
 
H
 et al.  
MiR-10a rejuvenates aged human mesenchymal stem cells and improves heart function after myocardial infarction through KLF4
.
Stem Cell Res Ther
 
2018
;
9
:
1
16
.

93

Bertero
 
T
,
Cottrill
 
KA
,
Annis
 
S
 et al.  
A YAP/TAZ-miR-130/301 molecular circuit exerts systems-level control of fibrosis in a network of human diseases and physiologic conditions
.
Sci Rep
 
2015
;
5
:
18277
.

94

Segura
 
AM
,
Frazier
 
OH
,
Buja
 
LM.
 
Fibrosis and heart failure
.
Heart Fail Rev
 
2014
;
19
:
173
185
.

95

Dixon
 
IMC
,
Wigle
 
JT.
 
Cardiac Fibrosis and Heart Failure: Cause or Effect?
 
Swizerland
:
Springer
;
2015
.

96

Luo
 
J
,
Qin
 
L
,
Zhao
 
L
 et al.  
Tissue-engineered vascular grafts with advanced mechanical strength from human iPSCs
.
Cell Stem Cell
 
2020
;
26
:
251
261.e8
.

97

Desai
 
M
,
Seifalian
 
AM
,
Hamilton
 
G.
 
Role of prosthetic conduits in coronary artery bypass grafting
.
Eur J Cardiothorac Surg
 
2011
;
40
:
394
398
.

98

Dahl
 
SLM
,
Kypson
 
AP
,
Lawson
 
JH
 et al.  
Readily available tissue-engineered vascular grafts
.
Sci Transl Med
 
2011
;
3
:
68ra9
.

99

Dahl
 
SLM
,
Blum
 
JL
,
Niklason
 
LE.
 
Bioengineered vascular grafts: can we make them off-the-shelf?
 
Trends Cardiovasc Med
 
2011
;
21
:
83
89
.

100

Hoerstrup
 
SP
,
Cummings
 
I
,
Lachat
 
M
 et al.  
Functional growth in tissue-engineered living, vascular grafts: follow-up at 100 weeks in a large animal model
.
Circulation
 
2006
;
114
:
I159
66
.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/journals/pages/open_access/funder_policies/chorus/standard_publication_model)