Regenerative Approaches to Spinal Disorders: The Emerging Role of Adipose-Derived Stem Cells in Intervertebral Disc Regeneration

  1. Home
  2. Articles

Regenerative Approaches to Spinal Disorders: The Emerging Role of Adipose-Derived Stem Cells in Intervertebral Disc Regeneration

   

Dr. Jeremy Pont1*, Prof. Pankaj Singh2, Prof. Vivek Pratap Singh3 and Knightly Patterson4

1Clinical Director, Townsville Spine and Sports Med, Australia. Research Scholar, Department of medical biotechnology, Nims University Rajasthan

2Associate Professor, Department of Neurology, NIMS Hospital, NIMS University

3Associate Professor, Department of Biotechnology, Institute of Allied Medical Science & Technology (NIAMST), Nims University

4Dept of Physiotherapy, Australian Catholic University

*Corresponding author: Dr. Jeremy Pont, Research Scholar, Department of Medical Biotechnology, Nims University Rajasthan

Citation: Pont J, Singh P, Singh VP, Patterson K. Comparative Study of Adipose-Derived and Bone Marrow-Derived Mesenchyme Stem Cells for Intervertebral Disc Regeneration in Patients with Degenerative Disc Disease. J Stem Cell Res. 6(2):1-19.

Received: July 25, 2025 | Published: August 15, 2025

Copyright© 2025 genesis pub by Pont J, et al. CC BY-NC-ND 4.0 DEED. This is an open-access article distributed under the terms of the Creative Commons Attribution-Non Commercial-No Derivatives 4.0 International License.,This allows others to distribute, remix, tweak, and build upon the work, even commercially, as long as they credit the authors for the original creation.

DOIhttps://doi.org/10.52793/JSCR.2025.6(2)-76

Abstract

Back pain lasting for long periods due to degenerative intervertebral disc changes is one of the main conditions affecting millions worldwide, imposing a huge socioeconomic condition. The conventional treatment regime, which includes medical therapy and spinal surgeries, almost always aims at relieving symptoms and therefore does not really cease the degenerative cascade. As regenerative medicine advances, biological strategies are increasingly considered viable ways of repairing disc structure and function. Among the different cell therapies, adipose-derived stem cells have gathered the buzz as a preferable candidate because of their source from abundant adipose tissue, high retrieval number, immunomodulatory activity, and their ability to differentiate into various lineages.

This article discusses the mechanistic and translational possibilities of ADSCs in intervertebral disc regeneration while focusing on their roles in suppressing inflammation and in extracellular matrix formation, thereby differentiating to form nucleus pulposus-like cells. We undertake a critical review of the current bioengineering methodologies that attempt to enhance ADSC viability and integration into the hostile avascular disc environment, such as 3D scaffolds, hydrogels, and exosome-based delivery systems.

Preclinical and clinical evidence is provided to demonstrate the efficacy, safety, and constraints of each technique. Although results from early-phase trials are promising, there still remain key barriers to translation such as donor variability, immune contiguity, and cell survival amidst the hypoxic and acidic conditions. Finally, we propose future prospects involving gene editing, 3D bioprinting, and AI-assisted cell tracking at the interface of basic science and clinical application.

This article presents ADSCs with a multidisciplinary view as one of the critical building blocks in an evolving paradigm of regenerative spinal therapeutics, holding the potential for conferring durable, biologically mediated recovery to patients suffering from degenerative disc disease.

Keywords

Adipose-Derived Stem Cells (ADSCs); Intervertebral Disc Degeneration (IVDD); Regenerative Medicine; Stem Cell Therapy; Nucleus Pulposus; Extracellular Matrix; Spinal Disorders.

Introduction

Intervertebral disc degeneration is a common cause of chronic low back pain and spinal dysfunction on a global scale, occurring with different degrees of frequency in approximately 80% of persons at certain points in their lives [1]. The condition has its genesis in the gradual disintegration of the extracellular matrix (ECM) of the intervertebral disc, thereby bringing about dehydration of the nucleus pulposus, structural instability, inflammation, and, eventually, nerve compression and pain [2]. Current treatments such as non-steroidal anti-inflammatory drugs (NSAIDs), physical therapy, spinal fusion, and total disc replacement mostly address symptoms that are unable to arrest or reverse the concurrent degenerate cascade [3,4].

Because of the limited regenerative capacity of the avascular intervertebral disc, cell-based therapies have emerged as an enticing alternative to try to restore disc structure and function. Among various kinds of mesenchymal stem cells (MSC), those derived from adipose tissue have received attention due to their easy accessibility and high cell yield, with low donor-site morbidity, and efficient immunomodulatory and regenerative attributes [5,6]. In contrast to bone marrow-derived MSCs (BMSCs), ADSCs are easily accessed via minimally invasive procedures, with greatly enhanced survival in the nutrient-deficient and acidic environment of the degenerated disc [7].

Pathophysiology of Intervertebral Disc Degeneration

The intervertebral disc degeneration is a progressive multifactorial state characterized by certain biochemical, structural, cellular, and biomechanical changes in the disc. It is considered a major factor in the causation of chronic low back pain and a global source of disability, and hence, it has been the focus of most biological and regenerative interventions [4,5]. Understanding the molecular and tissue cascade of degeneration would be highly useful in the design of stem cell-based approaches such as ADSC-based therapy.

Normal structure and function of the intervertebral disc

The intervertebral disc bestowed with fibrocartilage comprises three anatomically and functionally different components: the nucleus pulposus (NP), annulus fibrosus (AF), and cartilaginous endplates (CEPs). Each of these components contributes in its own fashion towards the distribution of loads, structural stability, and biomechanical flexibility of the spine [3,8].

Disc Component

Composition

Physiological Function

Nucleus Pulposus

Gelatinous core rich in proteoglycans (aggrecan), type II collagen, and water

Resists compressive forces; distributes axial loads

Annulus Fibrosus

Concentric lamellae of type I collagen in outer layers; type II collagen in inner layers

Provides tensile strength; limits radial expansion of NP

Cartilaginous Endplate

Hyaline cartilage with type II and X collagen, sparse chondrocyte-like cells

Facilitates nutrient and waste exchange between disc and vertebral body; anchors the disc

Table 1: Major Components of the Human Intervertebral Disc.

In healthy adults, the NP is highly hydrated (approximately 70–90% water) because of the osmotic pull exerted by sulfated glycosaminoglycans. Organized bundles of collagen in the AF enclose the NP, while the CEPs permit passive diffusion of nutrients and metabolic wastes—this is important because the disc itself is avascular [9].

Biochemical and molecular degenerative cascade

IVDD begins with molecular disturbances, primarily the loss of proteoglycans, which decreased hydration eventually collapses the NP height. Simultaneously, change from type II to type I collagen occurs, particularly in the NP, impairing its compressive functions and increasing brittleness [10]. Resident disc cells enter senescence and apoptosis, notwithstanding their natural reparative potential [11].

Chronic inflammation emerges in a degenerating disc, with pro-inflammatory cytokines attaining an elevated level, including interleukin-1β (IL-1β), tumor necrosis factor-alpha (TNF-α), and matrix metalloproteinases (MMPs). The condition speeds up ECM degradation and impairs cellular homeostasis [12].

These changes are not different. Crosstalk between matrix destruction and inflammatory signaling results in a vicious cycle of degeneration and pain sensitization. Also, MMPs and ADAMTS enzymes further degrade the collagen and aggrecan networks and impair the load-bearing function of the disc [7,6].

Mechanical and systemic contributions

Some biomechanical overloads caused by bad posture, trauma, or abnormal curvatures may help accelerate degeneration by causing microtears in the AF, which increases hydrostatic pressure in the NP, and end plates get damaged [15]. These mechanical insults also cause inflammation and upregulate catabolic enzymes, worsening tissue loss.

Systemic factors of risks such as aging, smoking, diabetes, obesity, and genetic polymorphisms further degeneration by limiting nutrient diffusion, instigating oxidative stress, and promoting SASP (senescence-associated secretory phenotype), thereby speeding the exhaustion of cells [9,16].

Clinical relevance and regeneration need

Clinically, IVDD causes chronic back pain, radiculopathy, reduction of disc height, annular tears, and endplate changes on MRI. Conservative treatment such as analgesics or physical therapy is symptomatic only. Surgical interventions of invasive nature like spinal fusion are reported to hasten degeneration of adjacent segments [10,4].

This clinical need has initiated the search for regenerative therapies acting on the causes rather than on the symptoms of IVDD. Out of these, the waste is with great potency regeneration, modulation of immunity, and ease of harvesting through a minimally invasive procedure [5,7].

Adipose-Derived Stem Cells (Adipocyte-Derived Stem Cells: Characteristics and Potential)

Adipose-derived stem cells (ADSCs), a variety of mesenchymal stem cells, can be isolated from adipose tissue by a nearly noninvasive technique like liposuction. It was first put forth by Zuk et al. in 2001. ADSCs have the capacity to undergo self-renewal and multipotent differentiation and exhibit considerable paracrine effects. This makes them very attractive to tissue engineers, especially due to the hostile avascular environment in which degenerate intervertebral discs form [11,12].

ADSCs provide both logistical and biological advantages over other MSC sources like bone marrow or umbilical cord, as they give much higher cell yields per milliliter of tissue, have low donor site morbidity, and have strong immunomodulatory profiles. Secondly, they can survive and perform optimally even in microenvironments of very low oxygen and acid levels, i.e., the conditions that prevail in NP within degenerative discs.

Comparative evaluation of MSC sources for disc regeneration

The table below offers a comparison of ADSCs in terms of several regenerative and clinical parameters against BMSCs and UC-MSCs:

Property

ADSCs

BMSCs

UC-MSCs

Harvesting Procedure

Liposuction (minimally invasive)

Bone marrow aspiration (invasive)

Umbilical cord collection at birth

Cell Yield per mL

500,000–2,000,000 cells

10,000–100,000 cells

Variable

Donor Site Morbidity

Low

Moderate

None

Immunomodulatory Capacity

High

Moderate

Very High

Differentiation into NP-like Cells

High

High

Moderate

Survival in Hypoxic Environment

Good

Moderate

Unknown

Clinical Translation Readiness

High (many trials ongoing)

Moderate

Low to Moderate

Table 3: Comparison of Mesenchymal Stem Cell Sources for Intervertebral Disc Therapy.

Source: Compiled from Liu et al. (2022), Ha et al. (2017), Rochette et al. (2020), and Xie et al. (2021).

Phenotypic characterization and differentiation capacity

Typically, ADSCs express MSC surface markers CD73, CD90, and CD105 and lack hematopoietic markers CD34 and CD45 [7]. Under suitable culture conditions, ADSCs are multipotent stem cells that may differentiate into mesodermal lineages, including osteocytes, chondrocytes, and adipocytes. Concerning disc regeneration, ADSCs are capable of differentiating into a nucleus pulposus-like phenotype, with expression of SOX9, aggrecan, and collagen type II after induction with hypoxia or growth factors such as TGF-β and BMP-7 [8,13].

Preconditioning ADSCs under hypoxic conditions or co-culturing them with NP cells enhances viability and the expression of ECM proteins in disc-like environments [9]. Their secretory products, which consist of growth factors, cytokines, and extracellular vesicles, have the ability to modify the inflammatory environment and promote repair there without the cells needing to full differentiate into disc cells.

Paracrine activity and secretome function

ADSCs exert profound therapeutic effects through their paracrine secretion rather than direct tissue incorporation. Their secretome includes a wide range of bioactive molecules that promote ECM synthesis, inhibit apoptosis, and modulate immune responses.

Source: Derived from Li et al. (2024), Rochette et al. (2020), and Liu et al. (2022).

Immunomodulation and survival advantage

Along with regenerative potential, they may be immune privileged, allowing for possible allogeneic transplantation. They inhibited T-cell proliferation, induced macrophage polarization from M1 to M2 phenotype, and downregulated pro-inflammatory mediators, all of which are critical in limiting the inflammatory component of IVDD [12,2].

The enhanced survival of ADSCs in low-glucose, low-oxygen, and acidic conditions that mimic the degenerated disc environment has been demonstrated through several in vitro and in vivo models [10,7]. Such survival advantages make ADSCs more promising candidates having a putative role for therapeutics than other types of MSCs in treating spinal degeneration.

Viability and longevity of ADSCs in degenerated disc environment

Transplanted cells must survive, integrate, and function by withstanding the harsh microenvironment of the degenerated disc for therapies in intervertebral disc regeneration to be successful. Being a harsh environment, it consists largely of hypoxia, low glucose availability, mechanical pressure, and acidic pH-that together with the other environmental factors render these conditions hostile for most cell types to live in [17,4].

ADSCs are often believed to be much better adapted for these conditions when compared to MSCs derived from bone marrow. Under in vitro culture, ADSCs are shown to remain proliferative and secrete important trophic factors (TGF-β, IGF-1) and resist apoptosis under disc-mimicking conditions [6,16]. Such resistance could be attributed to their higher baseline expression of anti-apoptotic proteins and autophagy-related genes, which contributes to an enhanced capacity to alleviate cellular stress.

It has also been described that the secretion of extracellular vesicles (EVs) with microRNAs (miRNAs) such as miR-140 and miR-21 by ADSCs exerted cytoprotective effects on the neighboring disc cells by promoting matrix homeostasis and suppressing inflammation even in the absence of cell engraftment (Li et al., 2024).

Genetic stability and tumorigenic risk

The genetic stability of transplanted cells is an important criterion for the safety of stem cell therapy. Long-term in vitro expansion (especially suboptimally) could increase the chance of chromosomal aberrations and subsequent spontaneous transformation. However, ADSCs have maintained more or less stable karyotypes and hence pose a low risk of malignant transformation when cultured under GMP conditions [12].

Unlike induced pluripotent stem cells (iPSCs), ADSCs are only multipotent and are therefore less likely to form teratomas. Standardized quality controls and batch validations are still necessary to guarantee clinical safety when the process changes from autologous to allogeneic usages [18]. Several early-phase clinical.

Clinical applications and early trial outcomes

Trials were conducted to assess the feasibility, safety, and effectiveness of ADSC-based therapies for discogenic low back pain. In a phase I study, Ha et al. (2017) showed that an intradiscal injection of autologous ADSCs together with hyaluronic acid is safe and well-tolerated, but pain scores (VAS) and disability indices (ODI) improved during the 12-month follow-up.

In a further study [10], noticed increased MRI T2 signal intensities and disc hydration after ADSC transplantations, which may be signs of early tissue recovery. Hence a proof of concept from these trials that ADSCs shall ameliorate symptoms and perhaps restore the structural function of the disc, though larger and longer-term studies must be conducted to confirm these results.

Along with other approaches, several combination therapies, such as ADSCs in conjunction with biomaterial scaffolds, exosome-rich preparations, or gene-modified versions of ADSCs, are under active investigation aimed at maximizing therapies [9,5].

Regulatory considerations and future challenges

Despite the promising results, many hurdles are yet to be dealt with before ADSC therapy enters the realm of standard care. Presently, the FDA and EMA classify stem cell therapies as ATMPs requiring apt measures for quality controls, traceability, and evidence of efficacy. This includes dealing with:

  • Variability from donor to donor, e.g., age, comorbidities, tissue quality
  • Variability from batch to batch in cell manufacturing
  • Standardized dosing and delivery protocols for cellular therapies
  • Ensuring long-term safety follow-up of transplantation events

Popular trends, for example, allogeneic off-the-shelf ADSC therapy, bioprinted disc scaffold, and AI-guided cell tracking, may lead to solutions to some of these translational barriers within the coming decade [16].

Mechanisms of Disc Regeneration by Adipose-Derived Stem Cells

A coordinated set of biological mechanisms renders adipose-derived stem cells therapeutically effective for intervertebral disc regeneration. Instead of a single function of the cells, the mechanism of action of ADSCs depends on a synergistic network of cellular activities that include differentiation, paracrine effects, immunomodulatory activities, and structural matrix repair. All of these mechanisms, in concert, promote the regeneration of degenerate disc tissue while suppressing the pathological factors responsible for discogenic pain.

ADSCs work with this microenvironment after they are transplanted into the cage space. This environment is one characterized by hypoxia, acidity, nutrient deprivation, and active inflammation processes. The main advantage of ADSCs is the natural ability to adapt to adverse microenvironments. Unlike many mesenchymal stem cells, ADSCs can survive and function under the low-oxygen, low-glucose conditions characteristic of the nucleus pulposus (NP). They find expression of survival and anti-apoptotic genes to their benefit, and their viability, as well as biology, persists under disc-mimetic stress according to a few studies [7,6].

Although ADSCs can differentiate into NP-like cells, especially when stimulated with certain growth factors such as transforming growth factor-beta (TGF-β) or bone morphogenetic proteins (BMPs), most therapeutic effects occur through paracrine signaling. ADSCs secrete a powerful cocktail of growth factors, cytokines, and extracellular vesicles (EVs) that exert their influence on neighboring cells, modulate the immune system, and stimulate extracellular matrix (ECM) synthesis [5,11]. These secreted molecules promote the survival and anabolic functions of resident disc cells, actively inhibit the production of catabolic enzymes, and suppress the pro-inflammatory cascade that causes pain and tissue degradation.

Their exosome-rich secretome takes a special stand. These EVs contain bioactive molecules, including microRNAs such as miR-21 and miR-140, which can reprogram the recipient disc cells, inhibit MMP activity, and induce the production of collagen type II and aggrecan. In contrast, ADSCs suppress pro-inflammatory cytokines such as TNF-α and IL-1β, while they up-regulate anti-inflammatory mediators such as interleukin-10 (IL-10), thereby providing a pro-regenerative ambience against chronic inflammation and fibrosis [12,16].

Being of low immunogenicity, ADSCs also induce immunotolerance upon the disc environment. They suppress T cell activity, promote macrophage differentiation toward the M2 anti-inflammatory phenotype, and reduce oxidative stress so that tissue healing occurs while also preventing immune-mediated rejection of the graft [9]. This property is invaluable, especially in allogeneic applications, where the host-versus-graft response is a major obstacle.

The multifactorial mechanism of action of ADSCs is illustrated in the diagram below, portraying how these cells impact myriad biological pathways to restore disc function.

Figure 3: Mechanisms of ADSC-Mediated Intervertebral Disc Regeneration.

Intertwined mechanisms acting together constitute an example of ADSCs acting not merely as cells for direct replacement but as bioactive mediators of tissue repair. Through molecular signaling, immune suppression, and matrix restoration, ADSCs can coordinate an entire regenerative microenvironment, making these stem cells uniquely equipped to deal with the complex pathology of intervertebral disc degeneration. This therapeutic profile represents a big advantage over single pharmacologic and surgical interventions and ultimately justifies further clinical investigations of ADSCs as the cornerstone of regenerative medicine in the spine.

Besides the aforementioned regenerative capacity, further factors influencing the efficacy of ADSCs include delivery method and biomechanical environment of implantation. These factors comprise conditions that hinder successful therapeutic intervention in the degenerated disc, such as high intracanal pressure, low vasculature, and disrupted extracellular matrix architecture. Being in opposition to one another, these factors are detrimental to not only the migration and retention of injected cells but also limit their potential to meaningfully interact with the native disc tissue. Consequently, even if ADSCs have such a rich secretome and plasticity, clinically, they will fail when having no chance of survival and activity for a significant time in the hostile disc environment [7,10].

In order to overcome such constraints, more recent reports have emphasized the persistence and activity of ADSCs, for example, co-delivery with supporting biomaterials such as hydrogels, alginate scaffolds, and thermoresponsive polymers. These delivery matrices are protective carriers, thus allowing a certain set of functionalities: increased cell retention, a controlled release of growth factors, buffering of pH, and distribution of mechanical load [9,14]. Some may even be bioactive in such a way as to provide cues for ADSCs to further develop into NP-like cells or to stimulate endogenous repair pathways. The amalgamation of ADSCs with these scaffolds certainly suggests an encouraging direction of work, as it can facilitate the synergistic interplay of biological and material sciences.

Genetic modification and preconditioning strategies, among others, present new attempts toward improving the therapeutic profile of ADSCs. For example, when ADSCs under hypoxic conditions are preconditioned, it is stated that they induce the expression of hypoxia-inducible factor-1α (HIF-1α), which further leads to secretion of vascular endothelial growth factor (VEGF) and stromal-derived factor-1 (SDF-1). The survivability and paracrine potentials of ADSCs thus initiated are better in vivo [5]. On the other hand, with an overexpression of anti-inflammatory cytokines or ECM-regulatory genes, ADSCs have been shown to possess superior therapeutic efficacy in preclinical disc regeneration models, indicating that advanced ADSCs can be tailored toward specific pathological features of IVDD.

Extracellular vesicles (EVs) derived from ADSCs are coming to the forefront as a cell-free alternate mode of therapy. EVs still provide many of the functional benefits associated with whole cell therapy, namely that of delivering microRNAs, anti-inflammatory molecules, and regenerative peptides, without declaring transplant-associated risks, such as ectopic tissue formation or immune sensitization. These vesicles can be grown on a large scale and can be stored and standardized, which turns into a big attraction for being a candidate for an off-the-shelf product for disc regeneration therapeutics [11,12].

A few unresolved issues remain concerning the translation of such promising findings. Especially, others noted the lack of consensus on therapeutic cells dosage, the variation in donor-derived ADSC potency, and even the absence of long-term clinical data on safety and long-term disc durability after transplantation. In addition, there should be ways to standardize the isolation, expansion, and quality control of ADSCs under a clinical-grade (GMP) scenario before approval and commercialization of such therapies can take place at an industrial scale.

Nonetheless, the biological versatility and mechanistic richness of the ADSCs have positioned them front and center in the regenerative strategies for IVDD. They do not stand alone but act as dynamic mediators capable of rewiring the disc microenvironment toward an anabolic, anti-inflammatory, and structurally reparative state. As research develops further, especially in bioprinting, exosome engineering, and real-time imaging of cell behavior in vivo, ADSC-based therapies shall advance from experimental interventions to real clinical treatments for millions suffering from degenerative spine diseases.

Bioengineering Strategies to Enhance ADSC Therapy

While the biological potential of ADSCs in the treatment of IVDD is clearly established, the reliance of clinical outcomes on the strategies employed to deliver, sustain, and optimize these cells is immense. In this case, peculiar physiological challenges ensue with the degenerated disc environment, impairing cellular viability, retention, and functional performance while granting low oxygen, acidic pH, poor vascular supply, and high intradiscal pressure. To surmount these obstacles, biomedical engineers conceived novel delivery systems and matrices to promote ADSCs' survival, integration, and regenerative activity in situ.

Hydrogel encapsulation is probably a very commonly explored technique for suspending ADSCs within a biocompatible matrix that emulates native nucleus pulposus in terms of mechanical and biochemical properties. Encapsulation has been performed with alginate-, hyaluronic acid-, and polyethylene glycol-based hydrogels. All these offers mechanical support while simultaneously providing a controlled-release activity for the secreted cytokines and growth factors. In addition, they help prevent cell leakage once injected into the disc space, which is a very typical problem in unprotected cell delivery [9,14].

This poses the establishment of 3-D bioprinting into a frontier for a domain. Layer-by-layer deposition of different biomaterials along with ADSCs allows researchers to manufacture disc-like constructs with spatial precision to recapitulate complex tissue architectures such as the NP-AF interface. This method theoretically enables preparing patient-specific disc grafts from autologous cells with customized geometry obtained from imaging data [5].

The nanofiber scaffold approach has also been producing promising results. These porous, ECM-mimetic structures-for the most part, polycaprolactone or collagen-can trap ADSCs within the disc space, align them, and stimulate matrix deposition. Alongside appropriate bioactive cues, such as integrin-binding peptides or gradients of growth factors, nanofiber scaffolds can exert even stronger therapeutic effects on ADSCs.

Additionally, preconditioning methods are used for improving ADSC survival and viability. In hypoxic preconditioning, hypoxia-inducible factors (HIFs) are expressed, and the secretion of pro-survival and regenerative factors like VEGF, IGF-1, and SDF-1 is increased. Once transplanted into the hypoxic environment of the disc, those preconditioned ADSCs have showed an enhanced capacity for survival, immunomodulation, and ECM production [6,7].

Genetic engineering approaches have also been employed to further increase the regenerative potential of ADSCs with overexpression of therapeutic genes, including those for TGF-β, BMP-7, or anti-inflammatory cytokines. These gene-modified cells show increased differentiation, ECM restoration, and inhibition of matrix-degrading enzymes like MMPs, yet the safety, regulatory, and ethical concerns currently pose a major impediment to the clinical translation of gene-modified ADSC therapies.

This is an innovative direction in exosome enrichment. Instead of whole-cell delivery, this technique exploits ADSC-derived extracellular vesicles (EVs) that convey regenerative miRNAs, cytokines, and proteins. Exosome therapy therefore circumvents issues related to cell viability, tumorigenicity, and immune rejection, while still providing strong paracrine signals that prompt endogenous repair mechanisms. Phase I trials and animal models have found exosomes to promote proliferation of disc cells, reduction of inflammation, and restoration of disc hydration [11,16].

Lastly, magnetic targeting approaches are increasingly seen as a novel approach to improve clinical level ADSC homing and retention. In this method, ADSCs are labeled with biocompatible magnetic nanoparticles and guided toward the degenerative disc region using external magnetic fields. Besides precise delivery, it curtails off-target diffusion and therefore may help reduce the required dose.

Strategy

Function

Hydrogel Encapsulation

Improves cell retention and mimics NP microenvironment

3D Bioprinting

Enables precise structural replication of disc architecture

Nanofiber Scaffolds

Provides mechanical support and promotes ECM synthesis

Hypoxic Preconditioning

Enhances paracrine factor secretion and cell survival

Genetic Modification (e.g., TGF-β overexpression)

Boosts regenerative gene expression and ECM remodeling

Exosome Enrichment

Delivers concentrated regenerative signals without live cells

Magnetic Targeting

Guides ADSCs to degenerated disc using external magnetic fields

Table 6: Bioengineering Strategies for Enhancing ADSC Therapy in Intervertebral Disc Regeneration.

Source: Adapted from Frith et al. (2013), Zhang et al. (2023), Liu et al. (2022), and Li et al. (2024).

Together, these bioengineering innovations are ushering in a transformation of ADSC therapy from simple cell injection to multimodal regenerative intervention. Interdisciplinary research combining materials science, cell biology, and clinical imaging aids the development of smarter, safer, and more targeted intervertebral disc repair strategies. These next-generation delivery systems not only enhance the therapeutic action of ADSCs but also serve as the fundamental starting point for personalized, scalable, and minimally invasive regenerative therapies for spinal disorders.

Preclinical and Clinical Data

There is a slow but steady accumulation of preclinical and clinical evidence in support of the journey of adipose-derived stem cells (ADSCs) from benchwork to bedside. Jointly, these studies prove the biological viability, safety, interest from regeneration, and effectiveness of ADSC therapies in intervertebral disc degeneration.

In the realm of preclinical research, animal models have come to represent a key consideration when seeking to experimentally confirm the mechanisms of ADSC action. For example, in a 2012 study [15], demonstrated that ADSCs injected in rabbit models of traumatic disc injury survived in the hostile disc microenvironment, formed part of the native tissue, and exerted beneficial effects on ECM synthesis and histological repair. In a similar fashion [5], have reported that, when encapsulated in a thermoresponsive hydrogel, ADSCs enhanced disc hydration and ECM restoration while decreasing levels of inflammatory cytokines against controls in the rat model of IVDD.

ADSC paracrine activity is also evident; multiple studies have reported increased anti-inflammatory cytokines (IL-10) and regenerative markers (aggrecan and collagen type II) after ADSC transplantation, therefore implying that ADSCs exercise their beneficial effects not only through direct cellular engraftment.

In terms of translation ability, early human trials have showed promising safety and efficacy data. Ha et al. (2017) applied a Phase I clinical trial involving intradiscal injection of autologous ADSCs combined with hyaluronic acid in 10 patients with chronic discogenic low back pain. The treatment was well-tolerated, with no serious adverse events, and showed statistically significant decreases in VAS and ODI scores at 12 months.

In a study involving 12 patients receiving intradiscal ADSC injections, either direct or indirect [10], corroborated these findings. Restoration of T2 signal intensity was observed on MRI post-treatment. This signified increased disc hydration, along with some possibility of structural restoration. Their own reports indicated improvement for pain and movement without signs indicative of discitis, neoplastic changes, or abnormal immune reactions.

A systematic review by [4], covering different cell therapy methods for treating back pain, including ADSC-based approaches, although the authors noted that the majority of trials reported clinically meaningful improvements in pain, function, and disc morphology, further stated the need for larger randomized controlled trials (RCTs), standardized cell preparation protocols, and longer-term follow-up to validate these preliminary findings.

Source: Compiled from peer-reviewed studies in Stem Cell Research & Therapy, Spine, and Tissue Engineering journals.

Preclinical studies in animals and some clinical trials demonstrated dramatic efficacy of ADSCs in the regeneration of degenerated discs-on which basis relatively small but convincing human studies were performed on patients.

Although highly encouraging, the findings must be considered with academic caution. Most human trials to date are small, without randomization or a control arm, and with a relatively short follow-up period. On the other hand, variations in ADSC preparation such as autologous versus allogeneic origin, their passage number, and the type of matrix used for delivery can create huge problems in avoiding reproducibility of outcomes. Therefore, the clinical translation of ADSC therapy in disc regeneration is still in the early phase of exploration, with largescale multicentric RCTs being urgently needed.

That said, the congruence of animal and human data, particularly with regard to biological plausibility and clinical feasibility, will act as a powerful base for the advanced consideration of ADSCs as a primary therapeutic candidate in regenerative spine medicine.

Challenges and Limitations

Varying major challenges remain, constraining the full clinical translation, despite the rapidly accumulating evidence pointing to the regenerative potential of adipose stem cells (ADSCs) in intervertebral disc degeneration (IVDD). Such limitations range from biological constraints to technical, regulatory, and logistical concerns—all issues that must be addressed at the very least since ADSC-based therapies could eventually be classified as routine care.

Another challenge is the retaining of cells after transplantation. The intervertebral discs are a very harsh microenvironment-viewed from degenerated under: hypoxic, acidic, mechanical stresses coupled with a lack of vascular perfusion. ADSCs comparatively suffer less under hypoxic conditions than other mesenchymal stem cell types [6], however thus, whereas retaining a significant number of cells die through apoptosis or get displaced soon after delivery, this problem becomes more pronounced when the transplantation is without a scaffold to support the cells. Hence, injection of ADSCs as free suspensions renders inefficient therapy and produces inconsistent outcomes [9].

Donor variability represents yet another problem. The regenerative features of ADSCs are affected by the donor's age, his or her BMI, metabolic profile, and any developing comorbidities. For example, cells taken from elderly or diabetic donors generally show low proliferation and differentiation potential, while their secretory profiles differ [12]. This is especially alarming as, in the case of autologous therapies, the afflicted persons with severe IVDD—mostly middle-aged or older—may therefore have less potent stem cells for their personal use.

Adding to the problem is the absence of standardized cell isolation and preparation procedures. Variations in enzymatic digestion protocols, passage numbers in in vitro expansion, or storage conditions could affect cell viability and properties such as phenotype and function. Therefore, even though it is increasingly common to apply GMP standards to cell preparation for clinical use, there is still much variation between laboratories and, thus, a bottleneck remains for reproducibility and regulatory approval [6].

Though ADSCs seem immunoprivileged, rejection could indeed occur if the allogeneic cells are used. The immune system of the host could react even to minimal expressions of the major histocompatibility complex (MHC), specifically in sensitized patients (heads). This adds to the concerns of repeated or high-dose administrations that may increase the chances of allo-sensitization with collagen [2]. Cell-free options such as exosome therapy hold promise as workarounds but are still in their infancy in clinical exploration.

There are logistical and regulatory hurdles aside from the biological and immunological issues needing consideration. These include:

  • The high costs of manufacturing, especially for autologous therapies requiring individualized processing.
  • Cold chain storage requirements, which pose problems for cell transport and scale-up.
  • Uncertain reimbursement scenario, wherein commercial deployment is financially uncertain.

Long-term safety surveillance has several gaps, as most of the trials being conducted presently report follow-up data only up to a few months.

Mostly, the theoretical concern of oncogenic transformation has been discussed as a genuine risk only when ADSCs are expanded extensively in vitro or turned genetically [18]. In cases of bone marrow mesenchymal stem cells, Kim and Im (2009) also describe tumorigenesis occurring from uncontrolled differentiation or ectopic tissue formation as a biological risk, especially if cells are not tightly regulated after delivery.

This medicine translation step is complicated clinically by the variability of outcomes seen in different trials. Patient selection, stages of disease included, method of delivery, and endpoints (e.g., pain scores versus imaging biomarkers) vary considerably across studies, making comparison difficult and consensus definition of protocols for therapeutic use elusive [10]. Furthermore, the lack of large double-blind RCTs makes drawing definite conclusions about efficacy and safety challenging.

Thus, while, in principle, ADSCs could offer enormous potential as regenerative treatment for IVDD, their successful implementation will have to be a multilateral effort encompassing basic science, translational research, clinical trials, manufacturing, and profile regulation. Addressing those challenges would be indispensable not only to validate therapeutic outcomes but also for setting up an actual scalable, safe, and cost-effective regimen for stem cell–based spine care.

Future Perspectives

As medicine progresses, particularly on the regenerative side, the role of ADSCs in spinal therapy is gradually translating from experimental intervention to translational promise. Now achieving this full potential clinically requires there being not only limitations removed but also an acceptance of strategic innovations in cellular engineering and delivery, manufacturing workflows, and regulatory oversight.

Among the most promising directions is next-generation cell delivery systems that maximize therapeutic retention and efficacy. Hydrogels, magnetic targeting, and 3D scaffolds have shown great promise in animal studies and in early clinical trials to better localize transplanted cells and augment their survival and function. These platforms may one day act as an active carrier that responds to triggering events in the disc, for example, changes in pH or mechanical stress, that cause the release of the ADSCs or the secretome in a controlled fashion, in response to the progression of the disease.

With donor variability being a main reason, the trend toward the establishment of curated allogeneic cell banks has become more apparent. These are supported by rigorous potency assays and donor profiling. - The banks could provide an "off-the-shelf" ADSC product that is immunocompatible, standardized, and ready for use, like current blood or bone marrow registries-growing processes. Meanwhile, parallel studies in cell reprogramming and hypoimmunogenic engineering are in the process of finding a way to create universal donor ADSCs that can completely circumvent immune detection by the host.

The scope of safety and logistics is increasingly exploring cell-free strategies of ADSC-derived extracellular vesicles (EVs). These biological nanoparticles allow several therapeutic effects of living cells, such as anti-inflammatory, anti-apoptotic, and matrix-restoring activity, but without the threat of random differentiation and immune sensitization. Not only that, but an EV-based product holds promises for easy storage, transportation, and scaling on a global market, thereby making it a strong contender commercially.

Establishing a clinical infrastructure is essential in creating patient registries, outcome measures, and trial design that are standardized and harmonized across studies. The regulatory agencies will have a central role in creating accelerated approval pathways for therapy development, whilst maintaining criteria equating to very high standards for safety, efficacy, and manufacturing integrity. An investment in automated bioprocessing technologies, which comply with good manufacturing practice standards, will further assure augmentation of production whilst driving down costs, contamination, and variability.

Customarily, developments in AI, bioinformatics, and regenerative medicine will overhaul, creating a new outlook for ADSC therapies. Predictive models could eventually model patient selection, dosing, and timing from an individualized pathology and biomarker profile-giving rise to personalized regenerative spine care.

The present challenges alongside forward strategies outlined previously are summarized below:

Source: Synthesized from Frith et al. (2013), Liu et al. (2022), Rochette et al. (2020), and Zhang et al. (2023).

With the fact in mind that ADSC therapy-the method-has its future in cultivating the capability to adapt in order to meet the multifactorial disc regeneration demand-technologically, clinically, and economically. In parallel with the advances in biomaterials, gene editing, AI diagnostics, and exosome sciences, ADSC-based therapies hold promise to completely change curing millions of patients with chronic spinal degeneration.

Conclusion

Intervertebral Disc Degeneration is considered one of the most prevalent degenerative spine disorders worldwide that is less amenable to therapeutic intervention, which generally furnishes symptomatic relief and no regenerative therapy in the long term. ADSCs thus present a biologically and clinically potent option for treating this pathology, with their regenerative potential including an ability to restore disc architecture, suppress inflammation, and block pain by interspersed cellular integration, paracrine action, and immunomodulation.

In the past two decades, stem cells, tissue engineering, and biomaterials advances have increasingly lent merit to ADSC applications. Preclinical studies have consistently demonstrated this to hold true at whatever the levels: disc preservation, restoring matrix, and downregulating catabolic signaling pathways. Early-phase clinical trials have basically proven ADSC interventions to be safe, feasible, and preliminarily efficacious with the additional support of hydrogel or exosome formulations.

The full therapeutic harnessing of adipose-derived stem cells (ADSCs) in spinal regeneration hinges upon transcending the really important translational barriers of donor variability, cell viability, and immune compatibility, along with manufacturing scale and pathways for regulatory approvals. Gene-enhancement of cells, scaffold-guided delivery of cells, enhancement by exosomes, along with automated bioprocessing systems, are all acting to mold the future framework within which these therapies might be deployed more safely, effectively, and economically.

Thus, more than just a novel intervention, ADSC therapy represents a true paradigm shift in biologically based, patient-specific care for degenerative spinal disease. It will be the continued collaborative research and well-construed clinical validation that will ensure that this promise is transformed into a durable clinical reality accessible to the world population at large.

References

  1. Wu PH, Kim HS, Jang IT. (2020) Intervertebral disc diseases part 2: A review of the current diagnostic and treatment strategies for intervertebral disc disease. Int J Mol Sci. 21: 2135.
  2. Ohnishi T, Homan K, Fukushima A, Ukeba D, Iwasaki N, et al. (2023) A review: Methodologies to promote the differentiation of mesenchymal stem cells for regeneration of intervertebral disc cells following intervertebral disc degeneration. Cells. 12(17): 2161.
  3. Wang YT, Wu XT, Wang, F. (2010) Regeneration potential and mechanism of bone marrow mesenchymal stem cell transplantation for treating intervertebral disc degeneration.  J Orthop Sci.  15:707-19.
  4. Wu T, Song HX, Dong Y, Li JH. (2018) Cell-based therapies for lumbar discogenic low back pain: Systematic review and single-arm meta-analysis. Spine. 43(1): 49-57.
  5. Zhang W, Wang D, Li H, Xu G, Zhang H, et al. (2023) Mesenchymal stem cells can improve discogenic pain in patients with intervertebral disc degeneration: A systematic review and meta-analysis. Front Bioeng Biotechnol. 11: 1155357.
  6. Liu ZH, Sun Z, Wang HQ. (2022) Application of stem cells in the repair of intervertebral disc degeneration: Preclinical evidence and future directions. Stem Cell Res Ther. 13: 339-50.
  7. Ha DH, Park JH, Shim JH, et al. (2017) Safety and tolerability of intradiscal implantation of combined autologous adipose-derived mesenchymal stem cells and hyaluronic acid in chronic discogenic low back pain: 1-year follow-up of a phase I study. Stem Cell Res Ther. 8 (1):262.
  8. Sakai D, Mochida J, Iwashina T. (2007) Adipose-derived stem cell therapy for intervertebral disc regeneration: Verification using in vitro reconstructed nucleus pulposus. Tissue Eng. 13(8):2085-95.
  9. Frith JE, Cameron AR, Menzies DJ, Ghosh P, Whitehead DL, et al. (2013) An injectable hydrogel incorporating mesenchymal precursor cells and pentosan polysulphate for intervertebral disc regeneration. Biomaterials. 34(37): 9430-40.
  10. Xie B, Chen S, Xu Y, Han W, Hu R, et al. (2021) Clinical efficacy and safety of human mesenchymal stem cell therapy for degenerative disc disease: A systematic review and meta-analysis of randomized controlled trials. Stem Cells Int.  Article ID 9149315.
  11. Li Y, Zhao W, Li W, Liu B. (2024) ADSC extracellular nanovesicles synergistic strategy for disc regeneration. Int J Mol Med, 54(3):  221-32.
  12. Rochette L, Mazini L, Malka G, Zeller M, Cottin Y. (2020) The crosstalk of adipose-derived stem cells, oxidative stress, and inflammation. Int J Mol Sci. 21(1): 234.
  13. Zhang Z, Li F, Tian H, Guan K, Guangmin Zhao, et al. (2014) Differentiation of adipose derived stem cells toward nucleus pulposus-like cells induced by hypoxia and a threedimensional chitosan-alginate gel scaffold in vitro. Chin Med J (Eng). 127(2): 314-21.
  14. Peroglio M, Grad S, Mortisen D. (2012) Injectable thermoreversible hyaluronan-based hydrogels for nucleus pulposus cell encapsulation. Eur Spine J. 21(6):839-49.
  15. Chun HJ, Kim YS, Kim BK, Kim EH, Kim JH, et al. (2012) Transplantation of human adipose-derived stem cells in a rabbit model of traumatic degeneration of lumbar discs. World Neurosurg. 78(4):364-71.
  16. Yan X, Wang X, Che Y, Feng P, Zhang T. (2025) Immunological mechanisms and immunoregulatory strategies in intervertebral disc degeneration. bioRxiv.
  17. Watanabe T, Sakai D, Yamamoto Y, Toru Iwashina, Serigano K, et al. (2010). Human nucleus pulposus cells significantly enhanced biological properties in a coculture system with autologous mesenchymal stem cells. J Ortho Res.28:623-30.
  18. Kim HJ, Im GI. (2009) Combination of transforming growth factor β2 and bone morphogenetic protein 7 enhances chondrogenesis from adipose tissue derived mesenchymal stem cells. Tissue Eng. Part A. 15(6):1543-51.
  19. Bellini E, Grieco M, Raposio E. (2017) The science behind autologous fat grafting. Ann Med Surg. 24:65-73.
  20. Chen S, Li K. (2019) Intervertebral disc regeneration with an adipose mesenchymal stem cell–TEC: A rat model study. J Tissue Eng. 10: 204173141983605.
  21. Collin EC, Grad S, Zeugolis DI, Vinatier CS, Clouet JR, et al. (2011) An injectable vehicle for nucleus pulposus cell-based therapy. Biomaterials, 32(12): 2862-70.
  22. Ely U, Smith T. (2020) Adipose derived stem cells: Current applications and future directions. Stem Cells Int. 8810813.
  23. Fang J, Chen F, Liu D, Gu F, Wang Y. (2021) Adipose tissue derived stem cells in breast reconstruction: Biology and translation. Stem Cell Res Ther.12:22.
  24. Humphrey JE, Reilly MJ. (2014) A systematic review of the safety and efficacy of mesenchymal stem cell therapy for intervertebral disc degeneration. Stem Cells Transl. Med, 3(3):354-63.
  25. Break MKB, Hussein W, Huwaimel B, Alafnan, A, Almansour K, et al. (2023) Artemisia sieberi Besser essential oil inhibits the growth and migration of breast cancer cells via induction of S-phase arrest, caspase-independent cell death and downregulation of ERK. J. Ethnopharmacol, 312: p.116492.
  26. Break MKB, Fung TY, Koh MZ, Ho WY, Tahir MIM, et al. (2023) Synthesis, crystal structure, antibacterial and in vitro anticancer activity of novel macroacyclic Schiff bases and their Cu (II) complexes derived from S-methyl and S-benzyl dithiocarbazate. Molecules. 28(13):5009.
  27. Li J, Sun Z, Zhao X. (2013) Adipose-derived stromal cells protect intervertebral disc cells from apoptosis under compressive load in vitro. Int J Biol Sci.  11(1):133-44.
  28. Mazini L, Ezzoubi M, Malka G. (2021) Overview of ADSC processing for therapeutic use: Pre- and post-COVID 19. Stem Cell Res Ther. 12:179.
  29. Risbud MV, Albert TJ, Guttapalli A. (2004) Differentiation of mesenchymal stem cells towards a nucleus pulposus like phenotype in vitro: Implications for cell-based transplantation therapy. Spine. 29: 2627-32.
  30. Strassburg S, Richardson SM, Freemont A J, Hoyland JA. (2010) Co-culture induces mesenchymal stem cell differentiation and modulation of the degenerate human nucleus pulposus cell phenotype. Regen Med. 5:701-11.
  31. Wei A, Tao H, Chung SA. (2023) Methodologies to promote differentiation of mesenchymal stem cells into intervertebral disc cells: A review. Cell. 12(17):2161.
whatsapp