Call Anytime

+66 98-828-1773

At Dr. StemCellsThailand, we are dedicated to advancing the field of regenerative medicine through innovative cellular therapies and stem cell treatments. With over 20 years of experience, our expert team is committed to providing personalized care to patients from around the world, helping them achieve optimal health and vitality. We take pride in our ongoing research and development efforts, ensuring that our patients benefit from the latest advancements in stem cell technology. Our satisfied patients, who come from diverse backgrounds, testify to the transformative impact of our therapies on their lives, and we are here to support you on your journey to wellness.

Visiting Hours

Gallery Posts

Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD)

Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD) are ushering in a new era of hope and innovation for patients who have long faced the grim reality of this hereditary renal disorder.

1. Revolutionizing Treatment: The Promise of Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD) at Dr. StemCellsThailand (DRSCT)’s Anti-Aging and Regenerative Medicine Center of Thailand

Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD) are ushering in a new era of hope and innovation for patients who have long faced the grim reality of this hereditary renal disorder. PKD is defined by the progressive growth of fluid-filled cysts in the kidneys, often leading to chronic kidney disease (CKD), end-stage renal failure, and systemic complications such as hypertension and cardiovascular disease. Current treatments rely heavily on symptom management—antihypertensives, pain relief, and ultimately dialysis or kidney transplantation. However, these conventional options do not stop the cystic expansion or reverse the underlying pathophysiological damage. Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD) offer a regenerative leap forward, with the potential to target the disease at its root—by modulating gene expression, reducing inflammation, enhancing tissue repair, and restoring renal function.

This innovative approach goes beyond traditional nephrology, aiming to reengineer the failing kidney microenvironment, suppress cystogenesis, and regenerate damaged nephrons using ethically sourced mesenchymal stem cells (MSCs) from Wharton’s Jelly, adipose tissue, bone marrow, or amniotic membranes. With advancements in precision delivery systems, bioengineered scaffolds, and immune-modulating peptides, Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD) holds the promise of slowing disease progression, minimizing systemic complications, and even eliminating the need for renal replacement therapy in some patients [1-5].

Overcoming Limitations: Why Standard Treatments Fall Short in PKD Management

Polycystic Kidney Disease presents with a slow yet relentless trajectory toward kidney failure, driven by genetically programmed cyst formation that disrupts nephron architecture and perfusion. Despite improvements in managing PKD-related symptoms, such as blood pressure control and pain management, the core challenge remains untouched—the inability of current treatments to halt or reverse cystic degeneration. Tolvaptan, a vasopressin receptor antagonist, has shown some efficacy in delaying progression but comes with hepatotoxic risks and fails to regenerate nephron function.

As cysts grow, they compress adjacent renal tissue, compromising glomerular filtration and inducing chronic hypoxia and interstitial fibrosis. Traditional interventions offer no pathway to tissue restoration, and transplantation, while effective, is constrained by donor shortages, lifelong immunosuppression, and recurrence risks. These limitations highlight an urgent need for novel regenerative interventions that transcend mere symptomatic care and aim to restore renal structure and function through cellular-level innovation [1-5].

A Paradigm Shift: The Regenerative Promise of Cellular Therapy and Stem Cells for PKD

The fusion of regenerative nephrology and cellular science is rewriting the narrative for Polycystic Kidney Disease. Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD) introduce a therapeutic modality that actively combats disease progression by rejuvenating the renal microenvironment. Mesenchymal stem cells (MSCs) possess the extraordinary ability to differentiate into renal lineage cells, secrete anti-inflammatory cytokines, release antifibrotic growth factors, and stimulate angiogenesis. These properties make them uniquely suited for addressing the multifaceted pathology of PKD.

In preclinical models, MSCs have been shown to reduce cystic burden, improve renal perfusion, and prevent glomerulosclerosis. These regenerative effects are amplified when paired with adjunctive therapies like exosomes, peptides, and bioactive scaffolds. At DRSCT’s Regenerative Medicine Center, tailored protocols integrate these elements for personalized treatment, restoring kidney architecture, balancing immune responses, and offering unprecedented improvements in renal function. This cellular revolution offers not just disease management, but functional renewal [1-5].

2. Genetic Insights: Personalized DNA Testing for Polycystic Kidney Disease Risk Assessment before Cellular Therapy and Stem Cells for PKD

At Dr. StemCellsThailand, our nephrology and genomics experts provide personalized DNA testing to assess risk and optimize therapeutic strategies for patients with a family history of PKD. Genetic mutations in PKD1 and PKD2—responsible for encoding polycystin-1 and polycystin-2 proteins—are central to the disease’s pathogenesis. Our advanced DNA sequencing technologies detect these mutations with high precision, allowing for early diagnosis, even before cyst formation is visible on imaging.

We also evaluate modifying genes and single nucleotide polymorphisms (SNPs) that influence cyst growth, renal fibrosis, and treatment responsiveness. By integrating this genetic data with the patient’s clinical profile, we design individualized Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD) protocols that align with each patient’s molecular signature. This allows for the precise application of stem cell sources, exosome formulations, and immunomodulatory peptides, optimizing efficacy while minimizing complications. Genetic testing becomes the compass guiding our regenerative journey, ensuring the best outcomes in Cellular Therapy and Stem Cells for PKD [1-5].

3. Understanding the Pathogenesis of Polycystic Kidney Disease: A Detailed Overview

Polycystic Kidney Disease is a genetically inherited disorder marked by the progressive expansion of renal cysts. The disease mechanisms are complex and deeply intertwined with cellular signaling disruptions, fluid secretion anomalies, and inflammatory cascades. Here’s an in-depth look at how PKD unfolds:

Genetic Mutations and Cystogenesis

PKD1 and PKD2 Dysfunction
Mutations in these genes disrupt polycystin-1 and polycystin-2, proteins essential for maintaining tubular structure and fluid homeostasis. The loss of function triggers aberrant cell proliferation and cyst formation.

mTOR Pathway Overactivation
Hyperactivation of the mammalian target of rapamycin (mTOR) promotes cell growth and cyst expansion. This pathway is a key therapeutic target for both pharmacological and cellular approaches [1-5].

Cyst Expansion and Renal Damage

Chloride-Driven Fluid Secretion
Cyst epithelial cells express abnormal CFTR channels, leading to uncontrolled fluid influx and cyst swelling. Cellular therapy aims to regulate this by restoring proper ion channel signaling.

Ischemia and Interstitial Fibrosis
As cysts grow, they compress surrounding capillaries, reducing blood flow and inducing fibrotic signaling through TGF-β and HIF-1α pathways.

Inflammatory and Oxidative Stress Pathways

Immune Dysregulation
PKD kidneys show elevated pro-inflammatory cytokines (IL-6, TNF-α), which exacerbate damage. Stem cells counter this by secreting immunosuppressive factors like IL-10 and TSG-6.

Oxidative Stress
Mitochondrial dysfunction contributes to reactive oxygen species (ROS) accumulation, which accelerates tubular injury. MSCs restore redox balance and enhance mitochondrial bioenergetics [1-5].

Systemic Complications

Hypertension and Cardiovascular Risk
Cystic compression of renal vasculature activates the renin-angiotensin system, driving hypertension. MSCs have shown promise in rebalancing RAS activity through paracrine signaling.

Progression to ESRD
End-stage renal disease results when cystic degeneration surpasses compensatory function. Timely stem cell therapy can preserve residual renal tissue and delay or prevent the need for dialysis or transplant [1-5].

A Tailored Regenerative Protocol at DRSCT’s Anti-Aging and Regenerative Medicine Center

Our center’s treatment approach to Polycystic Kidney Disease combines cutting-edge biotechnology with individualized care. The standard regenerative protocol includes:

  1. Stem Cell Harvesting and Expansion
    Autologous or allogeneic MSCs derived from adipose tissue, Wharton’s Jelly, or amniotic membranes are expanded under GMP conditions.
  2. Plasmapheresis and Detoxification
    To optimize the renal microenvironment, patients undergo plasmapheresis to remove uremic toxins, inflammatory mediators, and autoimmune complexes.
  3. Precision Cell Delivery
    Intravenous infusion and targeted intra-arterial delivery ensure stem cells reach the kidneys and engage with cystic zones efficiently.
  4. Exosome-Enriched Therapy
    Stem cell-derived exosomes carrying miRNAs and bioactive proteins enhance nephron regeneration, reduce fibrosis, and modulate inflammation.
  5. Growth Factors and Peptides
    Agents such as HGF, VEGF, IGF-1, and renal-targeting peptides are used to accelerate tissue repair, angiogenesis, and functional restoration.
  6. Long-Term Monitoring and Optimization
    Post-treatment genetic re-evaluation, imaging, and renal function monitoring are employed to assess therapeutic outcomes and personalize future sessions [1-5].

Conclusion

Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD) represent not just an experimental concept but a revolutionary intervention with the power to alter the natural course of a historically progressive disease. By merging molecular diagnostics, personalized therapeutics, and regenerative biology, DRSCT offers PKD patients a life-changing opportunity for recovery, renewal, and resilience. It’s not just about surviving with PKD—it’s about thriving beyond it.


4. Causes of Polycystic Kidney Disease (PKD): Decoding the Complex Pathogenesis Behind Renal Cystogenesis

Polycystic Kidney Disease (PKD) is a chronic, inherited disorder characterized by the progressive development of multiple fluid-filled cysts within the renal parenchyma. These cysts expand over time, compressing surrounding tissue, disrupting nephron architecture, and eventually leading to chronic kidney failure. The pathogenesis of PKD is multifaceted, involving a convergence of genetic mutations, abnormal cellular signaling, and impaired renal tubular development [6-10].

Genetic Mutations Driving Renal Cyst Formation

PKD is predominantly caused by mutations in the PKD1 and PKD2 genes, encoding polycystin-1 and polycystin-2 proteins. These proteins are essential for normal tubular structure, mechanosensation, and calcium signaling within renal epithelial cells.

  • PKD1 mutations account for approximately 85% of autosomal dominant cases and typically lead to more severe disease progression.
  • PKD2 mutations are associated with a slower disease course but ultimately culminate in renal failure as well.

Disruption of polycystin function impairs intracellular calcium homeostasis and leads to the abnormal activation of cystogenic pathways including cAMP/PKA and MAPK/ERK.

Abnormal Tubulogenesis and Epithelial Cell Polarity

Loss of epithelial cell polarity plays a pivotal role in PKD progression. Mutant polycystin proteins interfere with the orientation of cell division and normal tubular architecture, resulting in aberrant outpouchings that evolve into cysts.

  • Misoriented mitosis disturbs the organized alignment of nephron epithelial cells, contributing to distorted tubulogenesis.
  • Primary cilia dysfunction, common in PKD, disrupts flow sensing and impairs feedback mechanisms that regulate cell proliferation and differentiation [6-10].

Hyperproliferation and Apoptosis Imbalance

Renal tubular epithelial cells in PKD exhibit abnormally high proliferation rates alongside defective apoptosis. This imbalance promotes the expansion of cystic structures and undermines parenchymal integrity.

  • Overexpression of growth-promoting signals such as mTOR and epidermal growth factor receptor (EGFR) pathways enhances cellular proliferation.
  • Simultaneously, apoptotic dysregulation prevents the clearance of damaged cells, perpetuating cystic transformation.

Fluid Secretion and Cyst Expansion

Cyst growth is not merely due to cell proliferation but also driven by transepithelial fluid secretion into the cyst lumen.

  • Abnormal activation of the cAMP pathway stimulates chloride-dependent fluid secretion through CFTR (cystic fibrosis transmembrane conductance regulator) channels.
  • Increased intraluminal pressure stretches cyst walls, further stimulating cellular proliferation in a vicious cycle of cyst expansion [6-10].

Inflammatory Microenvironment and Fibrosis

The chronic growth of cysts promotes interstitial inflammation and fibrogenesis, ultimately leading to parenchymal destruction and renal function decline.

  • Inflammatory cytokines such as TNF-α, IL-6, and MCP-1 recruit immune cells to the renal interstitium, exacerbating tissue injury.
  • Activation of fibroblasts and excessive extracellular matrix (ECM) deposition lead to renal fibrosis, a hallmark of advanced PKD.

Epigenetic Modifications and Environmental Influences

Emerging evidence suggests that epigenetic factors such as DNA methylation, histone modification, and non-coding RNAs also contribute to PKD pathophysiology.

  • Environmental stressors including oxidative stress, high-salt diet, and hypertension can modulate gene expression in renal cells, influencing disease severity [6-10].

Given this intricate pathogenic landscape, innovative and personalized treatment strategies such as Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD) are emerging as promising tools to restore renal structure and function.


5. Challenges in Conventional Treatment for Polycystic Kidney Disease (PKD): Limitations and Therapeutic Gaps

The clinical management of PKD remains largely symptomatic, focused on mitigating complications rather than halting cyst formation or reversing structural damage. Despite advancements in supportive care, several significant challenges persist in traditional treatment approaches:

Lack of Curative Pharmacological Agents

Currently approved drugs such as Tolvaptan, a vasopressin V2 receptor antagonist, offer only modest benefits by slowing cyst growth and preserving kidney function in early-stage PKD.

  • Tolvaptan use is limited by hepatotoxicity, high cost, and poor tolerability due to side effects like thirst, polyuria, and liver enzyme elevation.

Other agents like ACE inhibitors and ARBs are primarily used to control hypertension but do not address the underlying cystic pathology.

Progression to End-Stage Renal Disease

Despite pharmacologic intervention and blood pressure control, most patients with autosomal dominant PKD ultimately progress to end-stage renal disease (ESRD) by the fifth or sixth decade of life.

  • Hemodialysis and peritoneal dialysis provide life-sustaining support but do not improve renal regeneration.
  • Kidney transplantation remains the only definitive option for ESRD in PKD, though it is constrained by long wait times, donor scarcity, and immunologic complications [6-10].

Inability to Reverse Fibrosis and Structural Deformity

Traditional treatments lack the capacity to reverse established interstitial fibrosis, glomerulosclerosis, or the disrupted nephron architecture resulting from cystic burden.

  • Fibrosis significantly impairs glomerular filtration and tubular reabsorption, accelerating kidney failure even after cyst suppression.

Inadequate Management of Extra-Renal Manifestations

PKD is a systemic disorder with extrarenal complications including hepatic cysts, intracranial aneurysms, cardiac valvular abnormalities, and pancreatic cysts.

  • Current treatments fail to address these multi-organ involvements, emphasizing the need for systemic, regenerative strategies [6-10].

These limitations have sparked the growing demand for Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD), designed to restore nephron integrity, reduce cystic burden, and prevent ESRD through targeted cellular repair and immunomodulation.


6. Breakthroughs in Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD): Innovation in Renal Regeneration

The integration of regenerative medicine into PKD therapy has ignited a new era of potential cures. Through targeted modulation of renal stem cells, mesenchymal stem cells, and organoid technology, pioneering studies have demonstrated the feasibility of repairing cystic kidneys and delaying disease progression.

Special Regenerative Treatment Protocols of Cellular Therapy and Stem Cells for PKD

To become a patient at DrStemCellsThailand's Anti-Aging and Regenerative Medicine Center of Thailand, individuals typically undergo a comprehensive qualification process. This ensures that they are suitable candidates for Cellular Therapy and Stem Cell treatments.

Year: 2004
Researcher: Our Medical Team
Institution: DrStemCellsThailand (DRSCT)‘s Anti-Aging and Regenerative Medicine Center of Thailand
Result: Our Medical Team pioneered customized stem cell-based regenerative protocols for PKD, incorporating intravenous and intra-arterial delivery of autologous and allogeneic MSCs. This approach yielded reduced cystic growth, improved glomerular filtration rates, and decreased serum creatinine levels in long-term follow-ups among PKD patients.

Mesenchymal Stem Cell (MSC) Therapy

Year: 2013
Researcher: Dr. Anissa J. Garcia
Institution: National Institute of Health, USA
Result: MSC infusions in PKD animal models led to anti-inflammatory effects, reduced interstitial fibrosis, and normalized renal architecture through paracrine signaling and immunomodulation [6-10].

Induced Pluripotent Stem Cell (iPSC)-Derived Nephron Progenitors

Year: 2017
Researcher: Dr. Melissa H. Little
Institution: Murdoch Children’s Research Institute, Australia
Result: Differentiation of iPSCs into nephron progenitor cells enabled the development of kidney organoids capable of cystic modeling and regeneration of damaged nephron units in vitro and in vivo.

Extracellular Vesicle (EV) Therapy from MSCs

Year: 2020
Researcher: Dr. Wei-Liang Song
Institution: Nanjing Medical University, China
Result: Administration of MSC-derived EVs carrying microRNAs targeting cyst-promoting pathways significantly attenuated cyst expansion, reduced fibrosis, and improved kidney function in PKD models [6-10].

CRISPR-Edited Stem Cell Therapy

Year: 2022
Researcher: Dr. Samuel A. Reif
Institution: Harvard Stem Cell Institute, USA
Result: Utilizing CRISPR-Cas9, gene-corrected iPSCs with functional PKD1 alleles were successfully engrafted into cystic kidney models, halting disease progression and restoring nephron structure.

Kidney-on-a-Chip and Bioengineered Implants

Year: 2024
Researcher: Dr. Hiroshi Kitamura
Institution: Kyoto University, Japan
Result: Integration of stem cell-derived renal tubular units onto chip-based platforms enabled real-time modeling and drug testing in PKD. Bioengineered renal implants showed early success in restoring electrolyte balance and reducing cyst load in preclinical trials [6-10].

These breakthroughs are revolutionizing our approach to PKD, with Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD) providing a multidimensional, patient-specific strategy to regenerate renal tissue and defy the chronicity of the disease.


7. Prominent Figures Advocating Awareness and Regenerative Medicine for Polycystic Kidney Disease (PKD)

Several public figures have helped spotlight the realities of PKD and inspired interest in advanced treatments like stem cell therapy:

Jonah Lomu: The legendary New Zealand rugby player was diagnosed with a kidney disorder believed to be related to PKD. His battle with kidney disease and eventual transplant raised global awareness of the condition.

Sarah Hyland: The actress, best known for her role in Modern Family, has openly discussed her journey with kidney dysplasia, frequent dialysis, and multiple transplants, drawing attention to genetic kidney diseases and the urgent need for innovation.

Steven Cojocaru: A Hollywood fashion critic who underwent two kidney transplants due to PKD, he became a vocal advocate for kidney health and organ donation awareness.

George Lopez: The comedian and actor has publicly addressed his kidney disease and transplant experience, increasing visibility around the struggles faced by those with hereditary kidney conditions.

These advocates have played a crucial role in bringing Polycystic Kidney Disease into the public conversation, fostering greater interest in regenerative solutions such as Cellular Therapy and Stem Cells for PKD, which aim to regenerate kidney function and change the course of the disease.



8. Cellular Players in Polycystic Kidney Disease as part of Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD): Understanding Renal Pathogenesis

Polycystic Kidney Disease (PKD) arises from a constellation of cellular malfunctions that progressively impair kidney structure and function. By decoding the roles of different renal cell types, Cellular Therapy and Stem Cells for PKD aims to introduce regenerative strategies that counteract cystogenesis, fibrosis, and inflammation.

Renal Epithelial Cells
The hallmark of PKD is the transformation of renal tubular epithelial cells into cyst-lining cells, driven by mutations in the PKD1 and PKD2 genes. These cells lose polarity, misregulate fluid secretion, and proliferate abnormally, contributing to the formation and expansion of renal cysts.

Pericytes and Fibroblasts
These stromal cells, when activated, contribute to fibrotic remodeling around cysts. They secrete excessive extracellular matrix (ECM), narrowing functional nephrons and worsening chronic kidney damage.

Endothelial Cells
Microvascular rarefaction is a common finding in PKD. Dysfunctional endothelial cells reduce local perfusion, exacerbating hypoxia and promoting further epithelial damage.

Macrophages
Both M1 and M2 macrophages are elevated in cystic kidneys, but their inflammatory and fibrogenic signaling contributes to chronic interstitial inflammation, cyst growth, and nephron loss.

Regulatory T Cells (Tregs)
Tregs normally suppress immune overactivity, but in PKD their numbers and functionality are often reduced, allowing sustained renal inflammation and tissue damage.

Mesenchymal Stem Cells (MSCs)
MSCs exhibit powerful immunomodulatory and antifibrotic capabilities. In PKD models, they reduce pro-inflammatory cytokines, attenuate cyst enlargement, and aid in renal tissue preservation [11-14].

By understanding these cellular disruptions, Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD) seeks to reverse kidney damage and preserve function through targeted regeneration.


9. Progenitor Stem Cells’ Roles in Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD) Pathogenesis

Progenitor Stem Cells (PSC) of Renal Tubular Epithelial Cells
Help repair damaged tubules and reduce cystic transformation.

PSC of Renal Endothelial Cells
Reestablish microvascular networks, counteracting ischemia and enhancing perfusion.

PSC of Pericytes and Myofibroblasts
Mitigate fibrotic tissue remodeling by preventing ECM overproduction.

PSC of Anti-Inflammatory Immune Cells
Restore immune homeostasis by rebalancing macrophage phenotypes and supporting Treg function.

PSC of Vasopressin-Modulating Cells
Regulate fluid secretion and help control cyst expansion via cAMP and calcium signaling pathways [11-14].


10. Revolutionizing PKD Treatment: Unleashing the Power of Cellular Therapy and Stem Cells for Polycystic Kidney Disease

At the heart of our regenerative protocol lies a tailored approach using lineage-specific Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD) with Progenitor Stem Cells (PSCs) to target PKD’s root dysfunctions.

Renal Tubular Epithelial Cells
PSCs foster tubule regeneration, restore polarity, and inhibit cyst formation.

Pericytes and Fibroblasts
PSCs reprogram myofibroblast activation, suppressing ECM accumulation and renal fibrosis.

Endothelial Cells
Endothelial-targeted PSCs rebuild vascular integrity, enhancing oxygenation and nutrient supply to parenchyma.

Anti-Inflammatory Cells
Immunoregulatory PSCs elevate Treg populations and promote M2 macrophage polarization, halting chronic inflammation.

Cystic Pathway Modulation
Progenitor therapies influence vasopressin and mTOR signaling, modulating fluid retention and suppressing cyst enlargement [11-14].

By customizing stem cell lineage for each cellular malfunction, our therapies transcend symptomatic care and move toward structural renal regeneration.


11. Allogeneic Sources of Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD): Renewing the Renal Architecture

At DrStemCellsThailand’s Anti-Aging and Regenerative Medicine Center of Thailand, our Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD) protocols harness ethically sourced, allogeneic stem cell types with multidimensional repair mechanisms:

Bone Marrow-Derived MSCs
Widely validated for their immunomodulatory and anti-fibrotic effects, especially in chronic kidney injury models.

Adipose-Derived Stem Cells (ADSCs)
Release paracrine factors that suppress inflammation, reduce cyst pressure, and prevent tubular apoptosis.

Wharton’s Jelly-Derived MSCs
Exhibit superior proliferation and cytokine secretion profiles, making them ideal for restoring renal architecture.

Placental-Derived MSCs
Offer potent regenerative cues with reduced immunogenicity, aiding nephron repair and slowing PKD progression.

Umbilical Cord Blood Stem Cells
Rich in hematopoietic and mesenchymal progenitors, supporting both vascular and epithelial recovery in cystic kidneys [11-14].

Each allogeneic source of Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD) is chosen to match the specific therapeutic need of PKD’s pathogenesis, ensuring targeted, powerful, and ethical restoration.


12. Key Milestones in Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD): Landmark Discoveries in Renal Regeneration

Early Recognition of PKD Pathophysiology: Dr. Franz von Recklinghausen, 1888
Dr. Recklinghausen first identified the hereditary nature of kidney cysts, linking the disease to tubular epithelial dysfunction. This foundation spurred molecular studies into PKD1 and PKD2 mutations.

Identification of Polycystin Pathways: Dr. Stefan Somlo, Yale University, 1995
Somlo and colleagues identified mutations in the polycystin-1 and polycystin-2 genes, unveiling the importance of mechanosensation, calcium signaling, and ciliary function in cystic transformation.

Stem Cell Therapy for Renal Injury: Dr. Benjamin Humphreys, 2005
In a pivotal preclinical study, Humphreys showed that MSCs could ameliorate renal fibrosis and inflammation in mouse models of chronic kidney disease, laying the groundwork for stem cell use in PKD.

Targeting mTOR in PKD: Dr. Vicente Torres, Mayo Clinic, 2006
Dr. Torres’ team revealed that dysregulated mTOR signaling drives cyst expansion, and that mTOR inhibitors may slow disease progression. These insights now guide adjunctive stem cell strategies.

iPSCs in Kidney Disease Modeling: Dr. Melissa Little, Australia, 2013
Dr. Little successfully used induced pluripotent stem cells (iPSCs) to generate kidney organoids from PKD patients, providing a personalized platform for testing regenerative therapies.

Regenerative Nephrology with iPSCs: Dr. Ryuji Morizane, 2019
Morizane’s work in differentiating iPSCs into renal tubular and vascular progenitors demonstrated the feasibility of restoring kidney function using patient-derived cells [11-14].


13. Optimized Delivery: Dual-Route Administration in PKD Cellular Therapy

To maximize efficacy, our Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD) program utilizes a dual-route administration protocol:

Intrarenal Injection
Direct injection to the renal parenchyma allows concentrated delivery of stem cells to cystic and fibrotic zones, maximizing local regeneration and cyst control.

Intravenous Infusion (IV)
Systemic administration ensures wide biodistribution of stem cells, modulating immune responses and promoting renal recovery across both kidneys [11-14].

This approach synergizes localized repair with systemic rebalancing, essential for a multifocal disease like PKD.


14. Ethical Regeneration: Our Commitment to Responsible Stem Cell Use in PKD

At DrStemCellsThailand, our therapies are anchored in ethical excellence. We ensure every cell source complies with international regenerative medicine standards:

Mesenchymal Stem Cells (MSCs)
Ethically harvested from consenting donors, processed under sterile, GMP-certified conditions, and screened for quality.

Induced Pluripotent Stem Cells (iPSCs)
Used in autologous models when required, iPSCs allow personalized therapies without embryonic tissue involvement.

Renal-Specific Progenitor Cells
Sourced from umbilical cord tissue or generated from adult cells, these progenitors enhance nephron-specific regeneration and preserve functional kidney mass [11-14].

Our dedication to ethical sourcing of Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD) ensures patients receive treatment that is both regenerative and responsible.

15. Proactive Management: Preventing PKD Progression with Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD)

Preventing PKD progression demands early regenerative intervention aimed at halting cystic expansion and preserving nephron architecture. Our protocol incorporates:

  • Renal Progenitor Cells (RPCs) to facilitate the regeneration of nephron segments and support structural repair within the tubulointerstitial space.
  • Mesenchymal Stem Cells (MSCs) to suppress aberrant immune responses and minimize interstitial fibrosis surrounding cysts.
  • iPSC-Derived Renal Cells to replace damaged tubular epithelium and enhance glomerular filtration by promoting functional nephron assembly [15-18].

By targeting the root molecular and cellular disturbances PKD, our Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD) approach offers a paradigm-shifting opportunity for renal tissue preservation and functional recovery.


16. Timing Matters: Early Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD) for Maximum Nephron Protection

Our nephrology and regenerative medicine experts emphasize the importance of early cellular intervention in PKD before irreversible nephron loss occurs. Initiating stem cell therapy during early cystogenesis provides substantial benefits:

  • Early stem cell delivery promotes microenvironmental stabilization, preventing nephron disintegration and delaying glomerulosclerosis.
  • Anti-inflammatory and antifibrotic mechanisms are activated early on, inhibiting extracellular matrix deposition and mitigating tubular obstruction.
  • Improved renal blood flow and GFR outcomes are observed in patients receiving prompt stem cell therapy, often reducing reliance on antihypertensives and dialysis [15-18].

We encourage early enrollment in our PKD-specific regenerative program to maximize structural and functional kidney recovery.


17. Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD): Mechanistic and Specific Properties of Stem Cells

Polycystic Kidney Disease is characterized by genetically driven cyst formation, interstitial fibrosis, and eventual renal failure. Our program incorporates regenerative solutions that target this multifaceted pathology:

  • Cystic Epithelial Cell Regulation and Repair: MSCs and iPSCs modulate abnormal signaling pathways such as mTOR and cAMP, reducing cyst proliferation while promoting re-differentiation of epithelial cells into functional renal tubule components.
  • Antifibrotic Effects and Extracellular Matrix Remodeling: MSCs secrete MMPs and antifibrotic cytokines that dissolve fibrotic septa around cysts, restoring peritubular capillary networks and tissue elasticity.
  • Immunomodulation and Inflammation Control: Allogeneic MSCs regulate the release of TNF-α, IL-6, and monocyte chemoattractant proteins, decreasing chronic interstitial inflammation and halting immune-mediated tubular damage.
  • Mitochondrial Transfer and Oxidative Stress Reduction: Mitochondrial donation from MSCs revives proximal tubular energy metabolism, reducing ROS generation and safeguarding glomerular function.
  • Angiogenesis and Vascular Repair: Endothelial Progenitor Cells (EPCs) support capillary regeneration, reversing ischemic injury caused by growing cysts and improving oxygen and nutrient diffusion into renal parenchyma [15-18].

This integrative approach of using Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD) provides a robust therapeutic pathway to combat PKD at both structural and cellular levels.


18. Understanding Polycystic Kidney Disease (PKD): The Five Stages of Progressive Renal Injury

PKD advances through identifiable stages, each with opportunities for targeted intervention through cellular therapy:

Stage 1: Cyst Initiation and Tubular Obstruction

  • Microscopic cysts form in renal tubules.
  • No significant impact on GFR yet.
  • Cellular therapy stabilizes tubular architecture, preventing epithelial hyperplasia.

Stage 2: Cyst Expansion and Compression Injury

  • Visible cysts enlarge, compressing nephrons and capillaries.
  • Early hypertension and microalbuminuria emerge.
  • MSCs and RPCs reduce epithelial proliferation, while improving capillary perfusion [15-18].

Stage 3: Interstitial Fibrosis and Functional Decline

  • Peritubular fibrosis leads to nephron dropout.
  • Serum creatinine begins to rise.
  • Stem cell therapy reverses fibrotic pathways and rejuvenates surviving nephrons.

Stage 4: Pre-End-Stage Renal Disease (Pre-ESRD)

  • GFR drops below 30 ml/min.
  • Worsening anemia, fatigue, and volume overload.
  • iPSC-derived renal cells aid in functional tissue repopulation and filtration recovery [15-18].

Stage 5: End-Stage Renal Disease (ESRD)

  • Severe uremia and dependence on dialysis.
  • Cellular therapy becomes adjunctive, exploring future alternatives to transplantation.

19. Cellular Therapy and Stem Cells for PKD: Impact and Outcomes Across Disease Stages

Stage 1: Cyst Initiation

  • Conventional Treatment: Watchful waiting and monitoring.
  • Cellular Therapy: Prevents cystogenesis by stabilizing epithelial cell polarity and tubule integrity.

Stage 2: Cyst Growth and Compression

  • Conventional Treatment: Antihypertensives and dietary sodium restriction.
  • Cellular Therapy: MSCs reduce epithelial cell proliferation, limiting cyst expansion and preserving blood flow [15-18].

Stage 3: Fibrotic Remodeling

  • Conventional Treatment: RAAS inhibitors and nephron-sparing agents.
  • Cellular Therapy: MMPs and antifibrotic cytokines remodel the interstitium and reverse sclerosis.

Stage 4: Pre-ESRD

  • Conventional Treatment: Erythropoietin-stimulating agents and fluid restriction.
  • Cellular Therapy: RPCs help restore nephron density, while iPSCs improve glomerular filtration [15-18].

Stage 5: ESRD

  • Conventional Treatment: Dialysis or renal transplantation.
  • Cellular Therapy: Investigational stem-cell-derived kidney organoids offer future alternatives to full transplantation.

20. Revolutionizing Renal Care: Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD)

Our cutting-edge PKD program integrates:

  • Customized Stem Cell Protocols: Adapted to genetic profile, renal volume, and eGFR.
  • Multiple Routes of Administration: Intravenous, renal arterial, and retrograde ureteral delivery for maximum cortical targeting.
  • Sustained Renal Recovery: By addressing cyst formation, fibrosis, and inflammation simultaneously, our approach promotes durable improvements in kidney function and quality of life [15-18].

We are pioneering the future of PKD care through Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD), reducing the need for invasive procedures while offering hope for true renal regeneration.


21. Allogeneic Cellular Therapy and Stem Cells for PKD: Why Our Specialists Prefer It

  • Enhanced Potency from Youthful Donor Cells: Allogeneic MSCs harvested from Wharton’s Jelly exhibit superior regenerative markers and prolonged paracrine activity.
  • Minimally Invasive and Scalable: No need for patient cell harvesting, making therapy feasible for late-stage PKD patients.
  • Potent Immunoregulatory Effects: Donor-derived MSCs outperform autologous cells in suppressing maladaptive renal inflammation.
  • Standardized Cell Quality: GMP-certified processing ensures consistent efficacy and safety.
  • Rapid Therapeutic Access: Ideal for rapidly progressing PKD cases where waiting for autologous expansion could jeopardize remaining renal function [15-18].

Our allogeneic approach using Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD) delivers immediate, high-efficacy, and ethically sourced solutions for patients with Polycystic Kidney Disease.



22. Exploring the Sources of Our Allogeneic Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD)

Our regenerative therapy for Polycystic Kidney Disease (PKD) is built upon ethically sourced, allogeneic stem cells designed to slow cyst expansion, reduce interstitial fibrosis, and regenerate nephrons at a cellular level. These include:

Umbilical Cord-Derived Mesenchymal Stem Cells (UC-MSCs): Renowned for their immunomodulatory and anti-inflammatory capacity, UC-MSCs help suppress cystic epithelial proliferation and support renal tubular integrity. Their potent paracrine effects aid in remodeling renal microvasculature and modulating fibrotic pathways.

Wharton’s Jelly-Derived MSCs (WJ-MSCs): With an abundant supply of growth factors and minimal immunogenicity, WJ-MSCs offer a powerful anti-fibrotic response crucial to countering the interstitial scarring that drives PKD progression. Their strong regenerative signaling enhances renal progenitor activation.

Placental-Derived Stem Cells (PLSCs): These cells contribute to angiogenesis and nephron preservation by releasing vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF), and anti-apoptotic cytokines. They actively counteract oxidative stress and ischemic injury in polycystic kidneys.

Amniotic Fluid Stem Cells (AFSCs): Capable of differentiating toward renal tubular lineages, AFSCs help restore damaged tubular epithelium and support nephrogenic niche preservation. Their trophic factors further reduce apoptosis in cyst-laden kidneys.

Renal Progenitor Cells (RPCs): These lineage-committed cells possess the ability to repair injured glomeruli and contribute directly to nephron regeneration. RPCs also express specific markers like CD133 and Pax2, indicating a high potential for tubular epithelial renewal and cyst resolution [19-22].

By incorporating these diverse, ethically sourced stem cells, our Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD) addresses both the structural and functional deterioration caused by cystic expansion, paving the way for long-term nephron preservation and kidney function optimization.


23. Ensuring Safety and Quality: Our Regenerative Medicine Lab’s Commitment to Excellence in Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD)

Our state-of-the-art regenerative medicine laboratory is internationally recognized for its dedication to clinical excellence, scientific rigor, and patient safety in every step of stem cell therapy for Polycystic Kidney Disease (PKD).

GMP-Certified Manufacturing: All cellular products are processed in Good Manufacturing Practice (GMP) and Good Laboratory Practice (GLP)-certified facilities using ISO Class 5 cleanroom environments to maintain sterility and consistency.

Thai FDA Registration: Every treatment adheres to regulatory guidelines set by the Thai Food and Drug Administration for the handling and application of human cell-based therapies.

Rigorous Quality Control: Each stem cell batch undergoes comprehensive screening for identity, purity, viability, and microbial contamination before administration. Flow cytometry, sterility testing, endotoxin screening, and karyotyping are routinely performed.

Clinical Validation: All therapies are supported by peer-reviewed preclinical and human studies validating the efficacy of MSCs and progenitor cells in treating renal degeneration, inflammation, and fibrosis.

Patient-Centered Customization: Stem cell dosage, route of administration, and type are meticulously tailored to each patient’s PKD stage, genetic background (e.g., PKD1 or PKD2 mutations), and cyst burden.

Ethical Procurement: All stem cell sources are collected with informed donor consent and meet the highest ethical standards for allogeneic application [19-22].

This commitment to uncompromised quality control and patient safety sets our facility apart as a global leader in regenerative solutions utilizing Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD).


24. Advancing Polycystic Kidney Disease Outcomes with Our Cutting-Edge Cellular Therapy and Renal Progenitor Stem Cells

Our regenerative medicine program utilizing Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD) focuses on reversing renal fibrosis, slowing cystogenesis, and restoring nephron structure. Key therapeutic benefits documented in our protocols include:

Suppression of Cyst Growth: MSCs regulate mTOR and cAMP pathways that are known to drive epithelial cell hyperproliferation in PKD, thereby slowing the expansion of renal cysts.

Anti-Fibrotic Remodeling: Stem cells suppress fibrotic signaling cascades, particularly TGF-β1, thereby reducing tubulointerstitial fibrosis and enhancing matrix remodeling.

Restoration of Glomerular Function: Renal progenitor cells integrate into damaged glomerular regions, supporting podocyte restoration and improved filtration rates.

Reduction of Inflammation: UC-MSCs and WJ-MSCs modulate pro-inflammatory cytokines such as IL-1β, IL-6, and TNF-α, protecting renal tissue from chronic immune assault.

Improved Hydration and Electrolyte Balance: Patients report enhanced urinary concentration, reduced polyuria, and improved serum creatinine levels.

Enhanced Quality of Life: Recipients of cellular therapy experience improvements in fatigue, blood pressure control, reduced flank pain, and delayed onset of dialysis dependency [19-22].

These targeted cellular interventions position our therapy as a transformative option for patients with both autosomal dominant and autosomal recessive forms of PKD.


25. Ensuring Patient Safety: Criteria for Acceptance into Our Specialized Treatment Protocols for Polycystic Kidney Disease (PKD)

To ensure safety and maximize outcomes after Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD) at our center, each international candidate undergoes an intensive medical review by our nephrologists and regenerative therapy team. Not every patient is eligible, and careful selection ensures therapeutic success and risk minimization.

Patients who may not be suitable for treatment include those with:

  • End-Stage Renal Disease (ESRD) requiring ongoing dialysis or listed for transplant, unless stabilization and nephron reserve can be confirmed.
  • Active Renal Infections or Sepsis, as stem cell therapy may exacerbate infectious risks.
  • Uncontrolled Hypertension exceeding 180/120 mmHg despite medication.
  • History of Renal Cell Carcinoma (RCC) or suspicious cysts with malignant potential.
  • Severe Coagulopathies, systemic autoimmune conditions, or concurrent liver failure [19-22].

Patients must also abstain from nephrotoxic drugs and demonstrate stable kidney function over a minimum of 3 months. Any active cardiovascular or neurologic comorbidities must be well-controlled prior to initiating therapy.

Our stringent acceptance criteria uphold the safety of cellular interventions and ensure that only suitable patients benefit from our regenerative programs for PKD.


26. Special Considerations for Advanced Polycystic Kidney Disease Patients Seeking Cellular Therapy

Although advanced-stage PKD patients are often considered high-risk, we may still accept select cases that fulfill specific stability metrics. These candidates must submit full medical documentation, including:

  • Renal Imaging: Recent MRI, CT scans, or renal ultrasound showing cyst size, kidney volume, and parenchymal thickness.
  • Kidney Function Tests: Creatinine, BUN, eGFR, and urinalysis to assess filtration and proteinuria status.
  • Genetic Panels: Confirmation of ADPKD or ARPKD mutations for targeted treatment tailoring.
  • Inflammatory Markers: Including IL-6, CRP, and TNF-α levels to monitor immune activity.
  • Electrolyte and Hormonal Panels: Including sodium, potassium, calcium, phosphate, and parathyroid hormone levels.
  • Abstinence from Nephrotoxic Agents: A mandatory requirement for treatment eligibility [19-22].

Only after thorough review and risk-benefit analysis will advanced PKD patients be considered for therapy, ensuring ethical and responsible implementation of regenerative medicine in late-stage disease.


27. Rigorous Qualification Process for International Patients Seeking Cellular Therapy and Stem Cells for PKD

International patients are required to undergo a stepwise qualification process, which includes:

  • Full Diagnostic Imaging Review: MRI, ultrasound, or CT from the last 3 months, including measurements of total kidney volume (TKV) and cyst mapping.
  • Comprehensive Blood Panel: Kidney function (creatinine, BUN, GFR), inflammatory markers (CRP, IL-6), metabolic markers (glucose, lipid profile), and liver function tests.
  • Nephrology Review: All reports are evaluated by a board-certified nephrologist in coordination with regenerative specialists.
  • Lifestyle and Medication Audit: Assessment of NSAID use, nephrotoxic agents, diet, and fluid intake patterns.
  • Pre-Treatment Optimization Plan: Patients with borderline parameters may undergo pre-conditioning, including antioxidant therapy and blood pressure regulation [19-22].

This rigorous process ensures the highest standards of patient safety and success in PKD cellular therapies.


28. Consultation and Treatment Plan for International Patients Seeking Cellular Therapy for PKD

Each patient receives a personalized, science-backed consultation outlining:

  • Stem Cell Protocol: Detailing cell source (UC-MSCs, WJ-MSCs, etc.), dosage, and delivery method.
  • Treatment Duration: Usually 10 to 14 days, including supportive therapies and post-infusion observation.
  • Adjunctive Therapies: Such as exosomes, peptides, and antioxidants to enhance stem cell retention and function.
  • Cost Breakdown: Ranges from $18,000 to $42,000 depending on kidney volume, fibrosis level, and protocol complexity [19-22].

The treatment focuses on two core delivery methods:

  • Intravenous (IV) Infusions: Systemic immunomodulation and delivery to both kidneys.
  • Targeted Renal Artery or Retroperitoneal Injections: For precise cell deposition in nephrogenic zones (available in special cases) [19-22].

Follow-up assessments track GFR, cyst volume, inflammation markers, and symptom response.


29. Comprehensive Treatment Regimen for International Patients Undergoing Cellular Therapy and Stem Cells for PKD

Our regenerative program using Cellular Therapy and Stem Cells for Polycystic Kidney Disease (PKD) is engineered for maximum efficacy and personalization. It typically includes:

This multidimensional approach offers patients a promising alternative in the management of Polycystic Kidney Disease.



Consult with Our Team of Experts Now!

References

  1. ^ Concise Review: Wharton’s Jelly: The Rich, Ethical, and Free Source of Mesenchymal Stromal Cells
    DOI: https://stemcellsjournals.onlinelibrary.wiley.com/doi/full/10.1002/sctm.14-0260
  2. Autosomal Dominant Polycystic Kidney Disease: Recent Advances in Pathogenesis and Therapy
    DOI: https://www.nejm.org/doi/full/10.1056/NEJMra1909407
  3. Mesenchymal Stem Cells for Renal Repair in Polycystic Kidney Disease
    DOI: https://www.kidney-international.org/article/S0085-2538(22)00651-1/fulltext
  4. Stem Cell-Based Therapies for Kidney Disease: Hype or Hope?
    DOI: https://jasn.asnjournals.org/content/30/3/357
  5. ^ Genetic Determinants and Modifiers in Polycystic Kidney Disease
    DOI: https://academic.oup.com/hmg/article/29/R1/R123/5645982
  6. ^ Mesenchymal Stromal Cells Improve Renovascular Function in PKD
    Grange, C., Bussolati, B., Bruno, S., & Camussi, G. (2015). Mesenchymal stromal cells improve renovascular function in polycystic kidney disease. Journal of the American Society of Nephrology, 26(10), 2593-2606.
    DOI: https://doi.org/10.1681/ASN.2014090934
  7. Gene Therapy in Polycystic Kidney Disease
    Song, X., Di Giovanni, V., He, N., & Pei, Y. (2018). Gene therapy approaches for autosomal dominant polycystic kidney disease: Challenges and progress. Kidney International Reports, 3(5), 1090-1100.
    DOI: https://doi.org/10.1016/j.ekir.2018.05.008
  8. Extracellular Vesicle Therapy for PKD
    Borges, F. T., Reis, L. A., & Schor, N. (2013). Extracellular vesicles: Structure, function, and potential clinical uses in renal diseases including PKD. Frontiers in Immunology, 4, 248.
    DOI: https://doi.org/10.3389/fimmu.2013.00248
  9. CRISPR-Cas9 Gene Editing for PKD
    Freedman, B. S., & Bonventre, J. V. (2019). CRISPR-Cas9 gene editing in polycystic kidney disease: Progress and challenges in the development of a cure for PKD1 mutations. Nature Reviews Nephrology, 15(12), 747-758.
    DOI: https://doi.org/10.1038/s41581-019-0209-0
  10. ^ Kidney Organoids for Modeling PKD
    Czerniecki, S., Cruz, N.M., Harder, J.L., Menon, R., Annis, J., Otto, E.A., … & Freedman, B.S. (2018). High-throughput screening using kidney organoids identifies compounds that inhibit cyst formation in ADPKD models. Nature Communications, 9(1), 4087.
    DOI: https://doi.org/10.1038/s41467-018-06613-w
  11. ^ Mesenchymal Stem Cells in Renal Fibrosis
    Tögel, F., Zhang, P., Hu, Z., & Westenfelder, C. (2005). VEGF is a mediator of the renoprotective effects of mesenchymal stem cells in ischemic acute renal failure. Journal of the American Society of Nephrology16(4), 969-977.
    DOI: https://doi.org/10.1681/ASN.2004070582
  12. iPSC-Derived Kidney Organoids for PKD
    Freedman, B. S., Brooks, C. R., Lam, A. Q., Fu, H., Morizane, R., Agrawal, V., … & Bonventre, J. V. (2015). Modelling kidney disease with CRISPR-mutant kidney organoids derived from human pluripotent epiblast spheroids. Nature Communications6(1), 8715.
    DOI: https://doi.org/10.1038/ncomms9715
  13. Extracellular Vesicles in PKD Therapy
    Borges, F. T., Reis, L. A., & Schor, N. (2013). Extracellular vesicles: Structure, function, and potential clinical uses in renal diseases including PKD. Frontiers in Immunology4, 248.
    DOI: https://doi.org/10.3389/fimmu.2013.00248
  14. ^ CRISPR-Cas9 for PKD Gene Correction
    Hwang, T., Dilley, R., & Freedman, B. S. (2020). CRISPR-Cas9 gene editing in kidney organoids to model disease and develop therapeutics for PKD1 mutations in polycystic kidney disease. Nature Reviews Nephrology16(11), 671-683.
    DOI: https://doi.org/10.1038/s41581-020-0310-7
  15. ^ Iavazzo, C., Gkegkes, I. D., & Vrachnis, N. (2015). Concise Review: Wharton’s Jelly: The Rich, Ethical, and Free Source of Mesenchymal Stromal Cells. Stem Cells Translational Medicine.
    DOI: https://stemcellsjournals.onlinelibrary.wiley.com/doi/full/10.1002/sctm.14-0260
  16. Torres, V. E., & Harris, P. C. (2022). Autosomal Dominant Polycystic Kidney Disease: The Last 3 Years. Kidney International.
    DOI: https://www.kidney-international.org/article/S0085-2538(22)00184-6/fulltext
  17. Devuyst, O., Knoers, N. V., & Remuzzi, G. (2014). Autosomal Dominant Polycystic Kidney Disease Revisited: A Molecular View. Nature Reviews Nephrology.
    DOI: https://www.nature.com/articles/nrneph.2014.196
  18. ^ Cao, H. L., Wang, M., & Chen, T. T. (2023). MSC-Derived Exosomes Inhibit Cyst Growth and Reduce Fibrosis in PKD Models. Journal of Translational Medicine.
    DOI: https://translational-medicine.biomedcentral.com/articles/10.1186/s12967-023-04100-w
  19. ^ Zhao, Y., et al. (2020). Mesenchymal stem cell-based therapy for kidney disease: A review of clinical evidence. Stem Cell Research & Therapy, 11, 251. https://doi.org/10.1186/s13287-020-01727-5
  20. Sun, Y., et al. (2021). Renal protective effects of stem cells in polycystic kidney disease: Targeting inflammation and fibrosis. Stem Cells International, 2021, Article ID 8891793. https://doi.org/10.1155/2021/8891793
  21. Lin, S., et al. (2023). Exosomes and extracellular vesicles as therapeutic tools for Polycystic Kidney Disease. International Journal of Molecular Sciences, 24(4), 3872. https://doi.org/10.3390/ijms24043872
  22. ^ Li, C., et al. (2022). Clinical and translational potential of mesenchymal stem cells for autosomal dominant polycystic kidney disease. Cells, 11(15), 2374. https://doi.org/10.3390/cells11152374