Stromal cells constitute a diverse and heterogeneous class of connective tissue cells that form the foundational infrastructure of organs and tissues throughout the body, providing essential structural support to parenchymal cells while facilitating key physiological processes such as tissuemorphogenesis, homeostasis, and regeneration.[1] These cells, often encompassing subtypes like fibroblasts, pericytes, telocytes, and mesenchymal stromal cells (MSCs), originate from mesenchymal precursors and are distributed across various tissues, including bone marrow, adipose tissue, skeletal muscle, skin, and lymphoid organs.[1][2]Characterized by their multipotent potential and dynamic interactions within the extracellular matrix, stromal cells play critical regulatory roles in modulating immune responses, supporting hematopoiesis, and promoting tissue repair through the secretion of bioactive factors with anti-inflammatory, anti-apoptotic, and immunomodulatory properties.[2] In the bone marrow niche, for instance, MSCs maintain hematopoietic stem cell quiescence and differentiation by expressing markers such as CD73, CD90, and CD105, while negatively lacking hematopoietic indicators like CD34 and CD45.[3][2] Their heterogeneity is evident in tissue-specific adaptations, such as CD34-positive telocytes in the gastrointestinal tract and skin that aid in myogenic support and wound healing, or fibroblastic reticular cells in lymphoid tissues that organize immune cell trafficking.[1]Beyond homeostasis, stromal cells significantly influence pathological conditions, including cancer progression where cancer-associated fibroblasts remodel the tumor microenvironment to enhance metastasis, and fibrotic diseases driven by dysregulated extracellular matrix production.[1] In regenerative medicine, their therapeutic potential is harnessed for applications like treating non-union fractures, graft-versus-host disease, and myocardial infarction, with clinical trials demonstrating improved outcomes through hMSC infusions that promote tissue repair and reduce inflammation.[2] This multifaceted nature underscores stromal cells' evolution from passive supporters to active orchestrators of tissue function and disease dynamics.[3]
Definition and Characteristics
Definition
Stromal cells, originally described in the 19th century as the cellular components of connective tissue derived from mesenchyme, form the supportive framework of various organs and tissues.[4] Early histological studies, such as those by Maximow in the early 20th century building on 19th-century observations, recognized their role in providing a microenvironment for cellular processes like hematopoiesis, though the term "stromal" gained prominence later with the identification of specific progenitor populations in bone marrow.[4]In modern biological usage, a key subset of stromal cells, mesenchymal stromal cells (MSCs), is defined by the minimal criteria established by the International Society for Cell & Gene Therapy (ISCT) in 2006.[5] These criteria emphasize their identity as non-hematopoietic, multipotent cells that provide structural and functional support to parenchymal cells—the primary functional cells of organs—through secretion of extracellular matrix components like collagen and fibronectin, as well as through paracrine signaling.[5][6] In culture, they exhibit plastic adherence and a fibroblast-like morphology, enabling expansion while maintaining self-renewal capacity.[5][7]The ISCT's 2019 position statement further clarifies the nomenclature, recommending "mesenchymal stromal cells" over "mesenchymal stem cells" unless rigorous in vitro and in vivo evidence demonstrates true stemness, as current definitions lack definitive proof of extensive hematopoietic or tissue reconstitution potential.[8] This distinction underscores their role as supportive progenitors rather than universal stem cells, prioritizing functional assays for therapeutic applications.[8]
Phenotypic Markers and Differentiation Potential
Stromal cells, particularly mesenchymal stromal cells (MSCs), are identified phenotypically through standardized surface marker expression profiles established by the International Society for Cell & Gene Therapy (ISCT). The minimal criteria require that at least 95% of the cell population express CD73, CD90, and CD105, while fewer than 2% express hematopoietic or endothelial markers such as CD11b, CD14, CD19, CD34, CD45, or HLA-DR, as verified by flow cytometry analysis.[5] These criteria, proposed in 2006 and reaffirmed in subsequent ISCT position statements, ensure the distinction of MSCs from other cell types by confirming their fibroblast-like properties and lack of lineage commitment.[5]A hallmark of stromal cell identity is their trilineage differentiation potential, assessed through in vitro assays that induce and confirm adipogenic, osteogenic, and chondrogenic lineages. For adipogenesis, cells are cultured in media supplemented with dexamethasone, insulin, indomethacin, and 3-isobutyl-1-methylxanthine for 2-3 weeks, followed by fixation and staining with Oil Red O to visualize intracellular lipid droplets as red inclusions under light microscopy.[9] Osteogenic differentiation involves exposure to β-glycerophosphate, ascorbic acid, and dexamethasone for 3-4 weeks, with calcium deposits detected by Alizarin Red Sstaining, appearing as orange-red nodules.[10] Chondrogenesis is induced in pellet cultures with TGF-β, dexamethasone, and ascorbic acid over 3 weeks, confirmed by Alcian Blue staining of glycosaminoglycans in the extracellular matrix, yielding blue coloration.[11] These protocols, integral to ISCT criteria, demonstrate the multipotency of stromal cells, though the efficiency and potency can vary depending on the tissue source.[5]Functional assays further validate stromal cell clonogenicity, with the colony-forming unit-fibroblast (CFU-F) assay serving as a key measure of proliferative potential. In this assay, single-cell suspensions are plated at low density (e.g., 10-100 cells/cm²) in standard culture media, incubated for 10-14 days, and stained with crystal violet to enumerate fibroblast-like colonies comprising at least 50 cells, indicating self-renewal capacity.[12] The trilineage differentiation remains the defining feature, but CFU-F frequency typically ranges from 1 in 10,000 to 1 in 100,000 nucleated cells in primary isolates, reflecting their rarity and underscoring the need for expansion in culture.[12]Recent research from 2024-2025 has identified additional markers to refine stromal cell subtypes, such as Sca-1 (stem cell antigen-1) in murine models, where high Sca-1 expression on MSCs correlates with enhanced metastatic support in breast cancer contexts and improved purification of adipose-derived cells with superior proliferative and differentiation potential.[13][14] These findings build on core ISCT markers by highlighting context-specific variations, particularly in preclinical studies.
Sources and Types
Primary Tissue Sources
Stromal cells, particularly mesenchymal stromal cells (MSCs), are primarily sourced from bone marrow, which serves as the gold standard due to its established role in supporting hematopoiesis.[15] In bone marrow aspirates, MSCs constitute approximately 0.001-0.01% of nucleated cells, yielding about 100-1,000 cells per milliliter of marrow.[16][17]Other key tissue sources include adipose tissue, which provides a higher yield of MSCs compared to bone marrow, with approximately 5,000 cells per gram of tissue or 0.35-1 million cells per gram, representing 1-10% of the stromal vascular fraction.[17][18]Umbilical cord and placental tissues are notable for their MSCs' high proliferative capacity and low immunogenicity, making them advantageous for allogeneic applications.[19][15] Additional sources encompass dental pulp, synovial fluid, and endometrium, each offering accessible MSCs with tissue-specific properties.[20][21][22]Isolation of MSCs from these sources typically involves density gradient centrifugation, such as using Ficoll to separate mononuclear cells, followed by selective plastic adherence in culture, where MSCs attach to the surface while hematopoietic cells are removed.[23][24] Yield comparisons highlight adipose tissue's superiority over bone marrow on a per-gram basis, facilitating larger-scale harvests for clinical use.[17][18]Quality of sourced MSCs is influenced by donor age, with older individuals exhibiting reduced proliferative potential and increased senescence markers, such as beta-galactosidase activity and shortened telomeres, leading to diminished therapeutic efficacy.[25][26] In contrast, placental-derived MSCs demonstrate enhanced "youthful" potency, effectively modulating aging-related neural pathways and preserving immunomodulatory functions in recent studies.[27] Differentiation potential can vary slightly by source, with adipose-derived MSCs often showing robust adipogenic capacity.[28]
Subtypes and Variants
Mesenchymal stromal cells (MSCs) represent the primary variant of stromal cells, characterized by their perivascular origin and close association with blood vessels, where they often manifest as pericytes or pericyte precursors.[29] These cells exhibit multipotency, differentiating into lineages such as osteoblasts, adipocytes, and chondrocytes, and are identified by their location in the vascular niche across various tissues.[30]Pericytes, as a key subset of MSCs, wrap around capillaries and contribute to vessel stability and homeostasis.[31]Specialized subtypes of stromal cells include cancer-associated fibroblasts (CAFs), defined as activated fibroblasts within the tumor stroma that express markers like α-smooth muscle actin and fibroblast activation protein.[32] Another distinct subtype is Sca-1high MSCs in murine models, a population highly expressing the stem cell antigen-1 (Sca-1) marker, identified in 2025 research as enriched in lung-derived MSCs with unique functional properties.[33]Muse cells, a pluripotent-like subpopulation of MSCs expressing SSEA-3 and capable of differentiating into cells of all three germ layers, show increased ratios under stress conditions, as demonstrated in 2025 studies on bone marrow-derived cells.[34]Environmental factors induce variants in stromal cells, such as hypoxia-adapted MSCs that upregulate pluripotency genes including Oct4 through hypoxia-inducible factor-2α (HIF-2α) activation, enhancing stemness under low-oxygen conditions.[35] Inflammatory-primed MSCs, exposed to cytokines like IFN-γ or TNF-α, exhibit a shifted secretome with altered production of immunomodulatory factors, promoting enhanced extracellular matrix modulation compared to unprimed cells.[36]Beyond core markers like CD73, CD90, and CD105, molecular distinctions define stromal cell variants; for instance, CD146 expression identifies pericyte-like subsets with vascular regulatory functions, while NG2 marks contractile pericytes involved in vessel tone control.[37] Placental sources yield variants with heightened immunomodulatory potential, though detailed origins are covered elsewhere.[38]
Physiological Functions
Support in Hematopoiesis
Stromal cells in the bone marrow microenvironment form critical architectural components of the hematopoietic niches, including the endosteal niche adjacent to the bone surface and the perivascular niche surrounding sinusoidal blood vessels. These cells, particularly mesenchymal stromal cells (MSCs) and CXCL12-abundant reticular (CAR) cells, provide a supportive scaffold that maintains hematopoietic stem cell (HSC) quiescence and facilitates their homing and retention through the expression of CXCL12 (also known as SDF-1). The CXCL12-CXCR4 signaling axis is essential for anchoring HSCs within these niches, preventing their premature differentiation or mobilization into the bloodstream.[39][40][41]Through paracrine signaling, stromal cells secrete key cytokines such as stem cell factor (SCF, also known as KIT ligand) and thrombopoietin (TPO), which promote HSC survival, self-renewal, and lineage commitment. SCF supports the proliferation and maintenance of HSCs and multipotent progenitors, while TPO regulates megakaryopoiesis and contributes to overall HSC pool stability. Additionally, stromal cells mediate direct cell-cell interactions via adhesion molecules like vascular cell adhesion molecule-1 (VCAM-1) and intercellular adhesion molecule-1 (ICAM-1), which bind to integrins on HSCs (e.g., VLA-4), enhancing their attachment and polarization within the niche to sustain quiescence and enable controlled differentiation. These mechanisms collectively form a physical and biochemical scaffold that balances HSC dormancy with the demands of blood cell production.[42][43][44][45]Experimental evidence from co-culture assays demonstrates that stromal cells significantly enhance the development of myeloid and natural killer (NK) cells from hematopoietic progenitors. For instance, human MSCs in co-culture with CD34+ cells promote myeloid differentiation through chemokine secretion and cell contact, leading to increased production of granulocytes and monocytes. Similarly, stromal support has been shown to drive NK cell maturation from early progenitors, with enhanced cytotoxicity observed in vitro. Recent advancements, such as 2025 models of artificial marrow organoids (AMOs), have recapitulated these interactions in a three-dimensional system, where MSCs efficiently support myeloid and NK lineage commitment from human induced pluripotent stem cell-derived progenitors, mimicking native bone marrow functionality.[46][47][48]Stromal cells form the non-hematopoietic compartment that regulates HSC dynamics. They modulate HSC self-renewal via Notch signaling, where stromal-expressed ligands like Jagged-1 and Delta-like 1 activate Notch receptors on HSCs, promoting their expansion and repopulation capacity without inducing exhaustion. This regulatory role ensures long-term hematopoiesis, with disruptions in stromal Notch components leading to impaired HSC maintenance in preclinical models.[49][50][51]
Role in Tissue Maintenance and Homeostasis
Stromal cells play a pivotal role in maintaining tissuearchitecture and function by synthesizing essential components of the extracellular matrix (ECM), including laminin and collagen types I and IV, which form basement membranes in various organs. In the lung, mesenchymal stromal cells produce these ECM elements to support alveolar septation, epithelial maturation, and overall structural integrity during homeostasis and repair processes.[52] Similarly, in the liver, hepatic stellate cells—a key stromal population—secrete collagen types I and IV along with laminin to reinforce basement membranes in the space of Disse, ensuring proper hepatocyte organization and sinusoidal patency under steady-state conditions.[53] These contributions prevent mechanical instability and facilitate nutrient exchange, underscoring stromal cells' foundational support for organ homeostasis.Beyond structural provision, stromal cells sustain parenchymal cell populations through targeted signaling interactions. In the gut, subepithelial stromal fibroblasts interact with Lgr5+ epithelial stem cells via Wnt ligands such as Wnt2b and amplifiers like RSPO3, driving crypt-villus axis renewal every 3–5 days to preserve mucosal barrier function and epithelial turnover. Recent 2025 studies on colonic stroma emphasize how these cells orchestrate Wnt/β-catenin pathway activation to maintain intestinal stem cell niches and prevent dysregulated proliferation during homeostasis.[54] This stromal-epithelial crosstalk exemplifies how non-hematopoietic support mechanisms ensure long-term tissue renewal across organs.Stromal cells also promote vascular homeostasis through angiogenic factors and pericyte functions. Pericytes, a specialized stromal subtype, secrete vascular endothelial growth factor (VEGF-A) in response to local cues, enhancing endothelial cell survival, migration, and capillary stabilization to maintain blood flow and prevent leakage.[55] By wrapping around vessels and modulating PI3K/AKT signaling, pericytes ensure microvascular integrity, which is crucial for nutrient delivery and waste removal in tissues like the lung and gut.In response to mild tissue injury, stromal cells engage feedback loops involving transforming growth factor-β (TGF-β) to coordinate repair while preserving homeostasis. TGF-β signaling in fibroblasts and pericytes induces controlled ECM remodeling, epithelial re-establishment, and angiogenesis without excessive deposition, relying on SMAD-mediated regulation of proliferation and apoptosis to restore balance.[56] This dynamic response highlights stromal cells' capacity to integrate injury signals for adaptive maintenance across multiple tissues.
Immunomodulatory Effects
Anti-inflammatory Mechanisms
Stromal cells, particularly mesenchymal stromal cells (MSCs), exert anti-inflammatory effects by suppressing effector immune cells through multiple pathways. These cells inhibit T-cell proliferation primarily via the production of indoleamine 2,3-dioxygenase (IDO), which depletes tryptophan essential for T-cell activation, and prostaglandin E2 (PGE2), which interferes with T-cell signaling and cytokine production.[57][58][59] Similarly, MSCs suppress B-cell antibody production and terminal differentiation by inducing cell cycle arrest and apoptosis through soluble factors like TGF-β and direct cell-cell interactions involving Fas/Fas ligand pathways.[60][61] For natural killer (NK) cells, MSCs modulate proliferation, cytotoxicity, and cytokine secretion, such as IFN-γ, by secreting IDO and PGE2, thereby reducing NK cell effector functions without inducing apoptosis.[62][63]In addition to effector suppression, stromal cells release soluble anti-inflammatory mediators that dampen immune responses. Key among these are transforming growth factor-β (TGF-β) and interleukin-10 (IL-10), which promote regulatory T-cell differentiation and inhibit pro-inflammatory cytokine production by macrophages and T cells. Stromal cell-derived extracellular vesicles (EVs) also contribute to anti-inflammatory effects by transferring bioactive molecules that modulate immune cell polarization and cytokine profiles, as demonstrated in recent studies on neurodegenerative and inflammatory diseases.[64][57][65] Upon stimulation with IFN-γ, MSCs upregulate inducible nitric oxide synthase (iNOS), leading to nitric oxide (NO) production that further suppresses T-cell proliferation and shifts macrophage polarization toward an anti-inflammatory M2 phenotype.[66][67]Cell-cell interactions also contribute significantly to the anti-inflammatory repertoire of stromal cells. MSCs express programmed death-ligand 1 (PD-L1) on their surface and secrete soluble PD-L1, which binds PD-1 on activated T cells, inducing T-cell anergy, exhaustion, and apoptosis while sparing regulatory T cells.[68][69] Recent studies have highlighted a novel mechanism involving neutrophil modulation, where MSCs induce neutrophil aggregation in the bone marrow, facilitating the release of extracellular vesicles that promote resolution of inflammation via immune rebalancing pathways and reduced systemic neutrophil activation.[70]The anti-inflammatory potential of stromal cells is highly context-dependent, often requiring priming with low levels of IFN-γ and TNF-α to optimize their secretome toward immunosuppressive factors like IDO, PGE2, and TGF-β, while higher cytokine concentrations can shift toward pro-inflammatory responses.[71][72]
Pro-inflammatory Mechanisms
Stromal cells, particularly mesenchymal stromal cells (MSCs), exhibit pro-inflammatory mechanisms when exposed to elevated levels of interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α), which serve as licensing signals to shift their phenotype toward immune amplification. This activation triggers intracellular pathways such as JAK/STAT1 for IFN-γ and NF-κB for TNF-α, leading to synergistic upregulation of pro-inflammatory cytokine production, including interleukin-6 (IL-6) and interleukin-8 (IL-8). These cytokines promote the chemotaxis and recruitment of monocytes to sites of inflammation, enhancing the innate immune response. Optimal licensing occurs with a 1:1 ratio of IFN-γ and TNF-α at approximately 60 ng/mL, followed by overnight exposure, which can double the immunomodulatory potency in peripheral blood mononuclear cell suppression assays while favoring pro-inflammatory effector functions.[73]In this pro-inflammatory state, stromal cells further support adaptive immunity by upregulating major histocompatibility complex class II (MHC-II) expression, which enhances their capacity for antigen presentation and interaction with T cells. This MHC-II induction, primarily driven by IFN-γ stimulation, allows stromal cells to modulate dendritic cell (DC) maturation, promoting a more immunogenic phenotype that presents antigens effectively to naive T cells. Concurrently, licensed stromal cells influence T helper cell differentiation, supporting Th1 and Th17 polarization in environments with activated CD4+ T cells by sustaining pro-inflammatory cytokine milieus like IL-6 and IL-12, thereby amplifying cell-mediated and humoral responses against pathogens. Such effects contrast with their baseline anti-inflammatory default but highlight their contextual adaptability in immune regulation.[66]Evidence for this dual role emerges from in vitro priming studies, where stromal cells undergo a phenotypic switch from immunosuppressive to pro-inflammatory states upon prolonged exposure to high-dose inflammatory cytokines, as demonstrated by increased secretion of chemokines and adhesion molecules like ICAM-1 and VCAM-1. Recent 2025 research on Sca-1^high stromal variants further illustrates this, showing their enhanced production of chemokines such as CCL2, CXCL1, and CCL7, which recruit pro-inflammatory myeloid cells including macrophages and neutrophils to amplify local inflammation during metastatic processes. These findings underscore the plasticity of stromal cells in responding to environmental cues.[67][13]This pro-inflammatory activation represents a transient phase in stromal cell function, facilitating initial pathogen clearance and immune cell orchestration before feedback mechanisms, such as prostaglandin E2 downregulation or IL-10 induction, promote resolution and tissue repair. By temporarily boosting effector responses, stromal cells ensure balanced inflammation without chronic escalation.[67]
Role in Pathology
In Cancer Progression and Microenvironment
Stromal cells within the tumor microenvironment, often termed tumor-associated stromal cells (TASCs), are recruited to tumor sites primarily through the stromal cell-derived factor-1 (SDF-1, also known as CXCL12)/C-X-C chemokine receptor type 4 (CXCR4) axis, where tumor cells or surrounding stroma secrete SDF-1 to attract CXCR4-expressing mesenchymal stromal cells from bone marrow or adjacent tissues, thereby facilitating their integration into the supportive tumor niche.[74] Once incorporated, TASCs promote tumor angiogenesis and cell survival by secreting vascular endothelial growth factor (VEGF) and interleukin-6 (IL-6), which stimulate endothelial cell proliferation and protect cancer cells from apoptosis under hypoxic or therapeutic stress conditions.[75][76]A prominent subtype of TASCs, carcinoma-associated fibroblasts (CAFs), particularly those expressing alpha-smooth muscle actin (α-SMA), actively drive epithelial-mesenchymal transition (EMT) in adjacent carcinoma cells via secretion of hepatocyte growth factor (HGF), which activates the c-MET receptor to enhance motility and invasive potential.[77] Additionally, α-SMA+ CAFs contribute to tumor invasion by remodeling the extracellular matrix (ECM) through elevated production of matrix metalloproteinases (MMPs) such as MMP-2 and MMP-9, which degrade basement membranes and create tracks for cancer cell dissemination.[78]In supporting metastasis, recent 2025 research has highlighted the role of Sca-1-high mesenchymal stromal cells (MSCs) in the lung, which recruit myeloid immune cells such as macrophages and neutrophils via CXCL1 secretion (among other factors) to precondition the pre-metastatic niche and enhance breast cancer colonization of pulmonary tissues.[13] Complementary 2025 machine learning analyses of single-cell RNA sequencing data from breast tumors have revealed significant stromal heterogeneity, identifying distinct CAF subpopulations that correlate with metastatic propensity through differential cytokine profiles, underscoring the need for subtype-specific targeting.[79]While stromal cells predominantly exhibit pro-tumorigenic functions in most solid tumors by fostering growth and immune evasion—such as through TGF-β-mediated suppression of T-cell infiltration—they can adopt anti-tumor roles when genetically engineered, for instance, to deliver interferon-β and inhibit tumor vascularization in preclinical models.[80][81] This immunomodulatory aspect briefly aids tumor immune evasion by dampening cytotoxic responses, though detailed mechanisms are addressed elsewhere.
In Inflammatory and Fibrotic Diseases
Stromal cells play a central role in the pathogenesis of fibrotic diseases by differentiating into myofibroblasts, primarily through the TGF-β/Smad signaling pathway, which drives excessive extracellular matrix (ECM) deposition and tissue scarring. In this process, TGF-β1 binds to its receptors on fibroblasts, activating canonical Smad2/3-dependent signaling that promotes myofibroblast transdifferentiation and the production of ECM components such as collagen and fibronectin. This mechanism is particularly evident in organ-specific fibrosis, where persistent activation leads to pathological remodeling; for instance, in liver cirrhosis, hepatic stellate cells—a type of stromal cell—undergo myofibroblast differentiation, resulting in excessive collagen deposition and progression to end-stage liver disease. Similarly, in idiopathic pulmonary fibrosis (IPF), alveolar fibroblasts contribute to lung stiffness through aberrant ECM accumulation, exacerbating respiratory failure.Beyond fibrosis, stromal cells amplify chronic inflammation in diseases like inflammatory bowel disease (IBD) by secreting pro-inflammatory cytokines such as IL-6, which promotes Th17 cell differentiation and sustains immune-mediated tissue damage. In the intestinal mucosa of IBD patients, fibroblasts upregulate IL-6 in response to Th17-derived signals like IL-17, creating a feedback loop that enhances Th17 responses and drives mucosal inflammation toward fibrosis. Recent studies on placental mesenchymal stromal cells (MSCs) have highlighted potential regulatory pathways, showing that activation of the AMPK-FXR axis in these cells can mitigate gut inflammation in Crohn's disease models by reducing pro-inflammatory cytokine release and restoring barrier integrity.[82]In rheumatoid arthritis (RA), synovial stromal cells, particularly fibroblasts, perpetuate joint inflammation and erosion by forming a pannus-like structure that invades cartilage and bone, with upregulated expression of stromal markers correlating to disease severity. These cells respond to inflammatory cues by secreting chemokines and cytokines that recruit immune effectors, maintaining chronic synovitis. In scleroderma (systemic sclerosis), dermal stromal cells, including LGR5-expressing fibroblasts, drive skin fibrosis through dysregulated ECM production and vascular alterations, leading to progressive skin thickening and internal organ involvement.Disease progression in these conditions is further exacerbated by persistent injury signals that induce stromal cell senescence, triggering the senescence-associated secretory phenotype (SASP) and amplifying inflammation and fibrosis. Senescent stromal cells, such as fibroblasts in fibrotic lungs or hepatic stellate cells, release SASP factors including IL-6 and TGF-β, which perpetuate myofibroblast activation and ECM deposition in a self-sustaining manner. This senescent state, often linked to unresolved tissue damage, contributes to the transition from acute injury to chronic fibrotic disease across multiple organs.
Therapeutic Applications
Regenerative and Tissue Repair Therapies
Stromal cells, particularly mesenchymal stromal cells (MSCs), play a pivotal role in regenerative therapies by leveraging both direct differentiation into tissue-specific lineages and paracrine mechanisms to promote repair. In direct differentiation, MSCs can integrate into damaged tissues and contribute to structural regeneration, such as forming chondrocytes for cartilage repair or osteoblasts for bone healing.[83] Paracrine effects are mediated through secreted factors, including growth factors like vascular endothelial growth factor (VEGF) and hepatocyte growth factor (HGF), which stimulate angiogenesis and wound healing in surrounding cells.[84] Additionally, MSC-derived exosomes carrying microRNAs (miRNAs), such as miR-126 and miR-21, enhance endothelial cell migration and proliferation to support vascular repair.[85]Clinical applications of stromal cells in tissue repair have advanced to phase III trials, demonstrating efficacy in specific conditions. For osteoarthritis, intra-articular injections of adipose-derived MSCs have shown significant improvements in pain scores and joint function, with a phase III randomized, double-blind trial reporting a mean WOMAC score reduction of 21.7 points at 6 months compared to 14.3 points in placebo (P = .002).[86] In myocardial infarction, bone marrow-derived MSCs administered intravenously post-infarct have improved left ventricular ejection fraction (LVEF) by an average of 4-6% in meta-analyses of randomized controlled trials, attributed to reduced infarct size and enhanced cardiac remodeling.[87]Recent advances in 2025 have focused on enhancing stromal cell potency for broader regenerative applications. Hypoxia preconditioning of multilineage-differentiating stress-enduring (Muse) cells, a pluripotent subset of MSCs, increases their yield by up to twofold and upregulates pluripotency genes like OCT4 and NANOG, enabling more efficient tissue regeneration in preclinical models of stroke and liver injury.[34]Organoid models incorporating stromal cells as supportive scaffolds have also progressed, with bone marrow-derived MSCs integrated into 3D structures to mimic hematopoietic niches and promote myeloid differentiation, offering scalable platforms for tissue engineering.[46]Despite these promising developments, challenges persist in stromal cell therapies, including low engraftment rates typically below 5% due to poor cellsurvival in hostile microenvironments and rapid clearance by the host immune system.[88] To address this, biomaterial scaffolds such as hydrogel-based matrices have been employed to improve cell retention and delivery, enhancing osteogenic potential in bone defect models by providing mechanical support and localized factor release.[89]
Immunomodulatory and Anti-inflammatory Therapies
Stromal cells, particularly mesenchymal stromal cells (MSCs), have been investigated in clinical trials for their immunomodulatory properties in treating autoimmune diseases, where they help suppress aberrant immune responses and reduce disease flares. In multiple sclerosis (MS), intravenous administration of autologous MSCs has demonstrated potential to attenuate symptoms by upregulating indoleamine 2,3-dioxygenase (IDO), an enzyme that inhibits T-cell proliferation and promotes regulatory T-cell expansion, leading to reduced relapse rates in phase I/II trials involving doses of 1-2 × 10^6 cells/kg. A phase I/II dose-finding study using human umbilical cord-derived MSCs in MS patients further confirmed safety and preliminary efficacy in modulating immune activity without significant adverse events. Similarly, in graft-versus-host disease (GVHD) following hematopoietic stem cell transplantation, MSCs have shown success in phases II and III trials by dampening donor T-cell reactivity and cytokine release, with overall response rates reaching 70% at 28 days post-infusion in steroid-refractory acute GVHD cases. As of 2025, regulatory efforts continue internationally, with the European Medicines Agency reviewing remestemcel-L for similar indications.[90]For acute inflammatory conditions, MSCs target excessive immune activation, such as cytokine storms. Clinical trials from 2020 to 2022 for acute respiratory distress syndrome (ARDS) associated with COVID-19 reported that intravenous MSC infusions reduced inflammatory biomarkers like IL-6 and TNF-α, improving oxygenation and lowering mortality risk in severe cases, as evidenced by a meta-analysis of randomized controlled trials showing significant decreases in cytokine levels and secondary outcomes like ventilator-free days. In Crohn's disease, placental-derived MSCs have emerged as a targeted therapy; a 2025 study demonstrated that these cells alleviate intestinal inflammation and repair barrier function by secreting IGFBP-4, which activates the AMPK-FXR pathway to suppress pro-inflammatory signaling, resulting in clinical remission in refractory patients.Delivery of MSCs primarily occurs via intravenous infusion, leveraging their intrinsic homing ability to inflamed sites through interactions with chemokines like SDF-1 and adhesion molecules such as VCAM-1, which guide cells to areas of tissue damage and immune dysregulation. Standard dosing regimens range from 1 to 2 × 10^6 cells/kg body weight, administered as single or multiple infusions spaced 7 days apart, ensuring safety and feasibility across trials in autoimmune and inflammatory settings.Meta-analyses of MSC therapies in immunomodulatory applications indicate response rates of 50-70% in conditions like GVHD and ARDS, with complete responses in 25-30% of cases and improved overall survival, particularly when combined with standard treatments. By 2025, the U.S. Food and Drug Administration (FDA) approved Ryoncil, an allogeneic bone marrow-derived MSC product, for steroid-refractory acute GVHD in pediatric patients, marking the first such approval for an immunomodulatory stromal cell therapy and paving the way for broader indications in autoimmune disorders.