Exosomes provide communication between cells (Vitamin D is needed)

Exosome Introduction

Tiny particles discovered in 1983

Exosomes were discovered in 1983 when researchers observed small vesicles released from maturing blood cells. They are the smallest extracellular vesicles at 30-150 nanometers in diameter, classified alongside larger microvesicles and apoptotic bodies. Multiple exosome types exist, characterized by their cell of origin and cargo content.[^1][^2][^3][^4][^5][^6]

Circulation via many fluids: blood, lymph, etc.

Exosomes travel through the bloodstream as well as lymph, saliva, urine, and cerebrospinal fluid. Their small size and lipid membrane allow them to traverse entire body systems and reach distant organs—traveling distances measured in feet rather than inches. They can even cross the blood-brain barrier to communicate between the brain and peripheral tissues.[^7][^8][^9][^10][^11]

One type of cell can transmit Exsomes to another type

Exosomes need not originate from the same cell type as the recipient cell. They mediate communication between different cell types—for example, dendritic cell exosomes activate T cells, and adipocyte exosomes signal to liver cells.[^12][^13][^14][^15]

Exosomes function in both health promotion and disease processes.

They facilitate intercellular communication, transfer genetic material (mRNA, miRNA), modulate immune responses, and support tissue repair. Therapeutically, they show promise in regenerative medicine, cancer treatment, and neurological disorders, though tumor-derived exosomes can suppress immunity and promote disease spread.[^5][^14][^16][^17][^18][^19]

Exosomes are a "novel endocrine hormone" Exosomes are recognized as mediators of long-distance cellular communication similar to hormones and are called a "novel endocrine hormone" by the Endocrine Society. Like hormones, they regulate interorgan crosstalk and metabolic homeostasis. However, exosomes deliver complex "packages" of proteins and nucleic acids rather than single signaling molecules.[^14][^20][^21][^22]

T-cells can both produce and receive Exosomes

While distinct from T cells, exosomes interact with them functionally. T cells produce exosomes that regulate immune responses, while dendritic and other immune cell exosomes activate or suppress T cell function. Exosomes serve as communication vehicles that modulate T cell activity rather than functioning as T cell equivalents.[^15][^18][^19][^23][^24][^25] [^26][^27][^28][^29][^30]

References for this section: Perplexity AI, 11/25


What Are Exosomes - Perplexity AI 11/25?

Exosomes are tiny membrane-bound extracellular vesicles that function as cellular messengers, carrying biological information between cells. They range in size from 30 to 150 nanometers in diameter, making them roughly the size of lipoproteins but much smaller than cells themselves.[^1][^2]

- - Structure and Composition

Exosomes consist of a lipid bilayer membrane that encloses a diverse cargo of molecular components derived from their parent cells. This cargo includes proteins (such as adhesion molecules, growth factors, and enzymes), lipids (including cholesterol and ceramides), and nucleic acids (DNA, mRNA, and microRNA). Because of their size and composition, exosomes are classified as a subset of the broader category of extracellular vesicles.[^2][^1]

- - How Exosomes Are Formed

The formation of exosomes follows a specific biological pathway. Early endosomes undergo inward folding, creating specialized compartments called multivesicular bodies (MVBs). Within these MVBs, intraluminal vesicles accumulate. When the MVB fuses with the cell membrane (plasma membrane), these intraluminal vesicles are released into the extracellular environment as exosomes. This release occurs through a process called exocytosis, and exosomes are found in various biological fluids including saliva, blood, urine, and cerebrospinal fluid.[^1][^2]

- - Functions in Cell Communication

Exosomes serve as critical mediators of intercellular communication. They transfer a wide range of molecular cargo—including proteins, RNA, DNA, and lipids—between cells, enabling them to regulate various biological pathways in recipient cells. This communication system operates both locally (between nearby cells) and systemically (between distant organs). Notably, exosomes can cross the blood-brain barrier, enabling communication between the peripheral nervous system and the central nervous system.[^3][^4]

- - Role in Health and Disease

Exosomes contribute to numerous fundamental physiological processes, including neuronal communication, immune responses, organ development, and antigen presentation. However, they also participate in pathological conditions. In cancer, exosomes from tumor cells may show differences in characteristics and can encapsulate information useful for disease detection, serving as potential biomarkers. Exosomes are also implicated in cardiovascular disease, inflammatory disorders, viral infections, and neurodegenerative diseases.[^5][^1]

- - Therapeutic Applications

The therapeutic potential of exosomes has become a major focus of biomedical research. Stem cell-derived exosomes have demonstrated promise in regenerative medicine, particularly for treating cardiac diseases such as myocardial infarction by promoting angiogenesis and improving heart function. They show potential for treating neurological disorders including Parkinson's disease and Alzheimer's disease. Mesenchymal stem cell (MSC)-derived exosomes have been explored for osteoarthritis treatment, where they inhibit inflammatory factors and improve joint function. Additionally, exosomes are being investigated for cancer treatment, gene therapy, and immune modulation.[^6][^7][^8]

Because exosomes naturally transfer biological signals and can be engineered to carry therapeutic molecules, they represent a viable alternative to cell-based therapies for various medical conditions.


Association Between Exosomes and Vitamin D - Perplexity AI 11/25

Yes, there is a significant and multifaceted association between exosomes and vitamin D. Research has revealed several important connections between vitamin D signaling, exosome production, and their biological functions.

- - Vitamin D Modulates Exosome Composition and Release

Vitamin D directly influences the content and characteristics of exosomes released by various cell types. Studies have shown that vitamin D enriches ceramide content in exosomes released by embryonic hippocampal cells, which plays a crucial role in cell differentiation. Specifically, vitamin D3 induces neutral sphingomyelinase activity, leading to the release of ceramide-enriched exosomes that can stimulate cell differentiation in neighboring cells.[^1][^2][^3]

In adipocytes, vitamin D modulates the lipid composition of exosomes, particularly affecting the membrane fluidity and budding processes. When vitamin D is present alone or under inflammatory conditions, it creates a specific lipidomic signature in both small and large extracellular vesicles, reinforcing its anti-inflammatory effects.[^4][^5]

- - Vitamin D Influences Exosomal microRNA Content

Recent research demonstrates that vitamin D regulates the microRNA (miRNA) content within exosomes. In adipocyte-derived extracellular vesicles, vitamin D modulates the expression of inflammatory microRNAs, representing a novel mechanism by which micronutrients regulate inflammation. In polycystic ovary syndrome (PCOS) models, vitamin D treatment significantly affected the expression of exosomal miR-186, miR-21, and various long non-coding RNAs (lncRNAs) including H19, ROR, and MALAT1. These findings suggest vitamin D plays a regulatory role in exosomal genetic cargo involved in disease pathogenesis.[^6][^7][^8]

- - Vitamin D Receptor and Exosome-Mediated Drug Resistance

The vitamin D receptor (VDR) is intimately connected to exosome function in disease processes. In hepatocellular carcinoma, exosomes derived from drug-resistant cancer cells can transfer resistance to parental cells, but vitamin D treatment effectively reverses this exosome-mediated drug resistance. Vitamin D pretreatment and co-treatment can prevent the transmission of aggressive phenotypes through exosomes and restore sensitivity to therapeutic agents like everolimus.[^9][^10][^11]

Interestingly, research on astronauts' plasma-derived exosomes revealed that spaceflight conditions induce aberrant epigenetic regulation of the vitamin D receptor through increased H3K27me3 marks, mediated by the enzyme EZH2. This suggests exosomes can carry signals that suppress VDR expression, potentially contributing to cardiovascular risks.[^12][^13]

- - Anti-Inflammatory Effects Through Exosome Modulation

Vitamin D's anti-inflammatory properties are partially mediated through its effects on exosomes. In macrophage-hepatic stellate cell communication, activating the VDR alters the protein profiles of exosomes derived from M2 macrophages and reverses their role in promoting hepatic stellate cell activation. This demonstrates that vitamin D can reprogram exosome function to reduce fibrosis and inflammation.[^14]

In vitamin D receptor-deficient keratinocytes, exosomal miR-4505 promotes macrophage polarization toward the pro-inflammatory M1 phenotype, contributing to psoriasis pathogenesis. This indicates that adequate vitamin D/VDR signaling is necessary to prevent the release of pro-inflammatory exosomes.[^15][^16]

- - Therapeutic Implications

The vitamin D-exosome connection has important therapeutic implications. Vitamin D treatment reduces inflammatory markers such as interleukin-8 (IL-8) in exosomes from PCOS models. The ability of vitamin D to modulate exosome content, composition, and function presents opportunities for developing novel therapeutic strategies that combine vitamin D supplementation with exosome-based therapies for conditions including cancer, metabolic disorders, inflammatory diseases, and cardiovascular conditions.[^17][^18]

In summary, vitamin D and exosomes are interconnected through multiple mechanisms: vitamin D modulates exosome lipid composition, influences their microRNA cargo, regulates their release, and affects how exosomes mediate intercellular communication in both health and disease states. [^19][^20][^21][^22][^23][^24][^25][^26][^27][^28][^29][^30][^31][^32][^33][^34][^35][^36][^37][^38][^39][^40][^41][^42][^43][^44][^45][^46][^47]


How Exosomes Support Human Health - Perplexity AI 11/25

Exosomes are nanosized extracellular vesicles—ranging from 30 to 150 nanometers in diameter—that play a crucial role in intercellular communication throughout the human body. Once considered merely cellular waste products, these tiny membrane-bound structures are now recognized as powerful biological messengers that orchestrate numerous physiological processes essential to human health. They carry a diverse cargo of bioactive molecules including proteins, lipids, growth factors, messenger RNA (mRNA), microRNA (miRNA), and other nucleic acids, which they transfer from donor cells to recipient cells.[^1][^2][^3][^4][^5]

- - Cell Communication and Regenerative Medicine

The primary function of exosomes is facilitating communication between cells by delivering specific molecular instructions that regulate cellular activities such as proliferation, differentiation, and migration. This intercellular signaling is fundamental to coordinating complex regenerative processes throughout the body. Exosomes derived from stem cells—particularly mesenchymal stem cells (MSCs)—have demonstrated remarkable regenerative potential by activating endogenous stem cells and promoting tissue repair without requiring direct stem cell transplantation.[^3][^6][^7][^1]

In regenerative medicine, exosomes contribute to tissue repair through multiple mechanisms including promoting angiogenesis (formation of new blood vessels), stimulating collagen production, and modulating immune responses. Research shows that stem cell-derived exosomes contain approximately three times as many growth factors as adult stem cells themselves, which enhances their ability to repair and revitalize target cells.[^6][^8][^3]

- - Orthopedic and Joint Health

Exosomes have shown significant therapeutic potential in treating musculoskeletal conditions, particularly osteoarthritis (OA) and joint injuries. MSC-derived exosomes can promote chondrogenesis (cartilage formation), reduce inflammation in joints, and improve joint function by modulating immune responses and delivering anti-inflammatory cytokines. In bone healing, exosomes enhance fracture repair by promoting both osteogenesis (bone formation) and angiogenesis, potentially through the BMP-2/Smad1/RUNX2 pathway.[^2][^9][^10][^11][^1][^6]

Studies demonstrate that exosomes can alleviate cartilage injury, reduce chondrocyte apoptosis, inhibit extracellular matrix degradation, and maintain cartilage homeostasis. When combined with biocompatible materials like hydrogels, exosomes facilitate cartilage tissue engineering therapies for osteoarthritis.[^12][^13][^14]

- - Neurological Health and Brain Function

Exosomes play a vital role in nervous system health, showing promise in treating various neurodegenerative conditions including Alzheimer's disease, Parkinson's disease, stroke, traumatic brain injury, and amyotrophic lateral sclerosis (ALS). MSC-derived exosomes administered systemically have been shown to significantly improve functional outcomes after ischemic stroke by enhancing angiogenesis, neurogenesis, and neurite remodeling in the brain.[^15][^16][^1][^2]

In stroke treatment, exosomes reduce apoptosis, modulate inflammation, stimulate cell proliferation, promote neurogenesis and synaptic plasticity, and deliver immunomodulatory signals. They can also reduce neuroinflammation, prevent reactive astrogliosis, and improve functional recovery in brain injury models. Importantly, exosomes can be engineered to cross the blood-brain barrier, enabling targeted delivery of therapeutic agents to the central nervous system.[^16][^17][^15]

- - Cardiovascular Protection

Exosomes contribute to cardiovascular health by promoting cardiac tissue repair and enhancing heart function following injury. Exosomes derived from stem cells and endothelial progenitor cells can stimulate angiogenesis, improve cardiac function after myocardial infarction, and enhance the survival and proliferation of cardiac progenitor cells. Their ability to modulate inflammatory responses and promote blood vessel formation makes them valuable for cardiovascular regenerative therapies.[^8][^18][^1][^2][^16]

- - Immune System Modulation

Exosomes serve as critical regulators of immune responses, capable of both stimulating and suppressing immunity depending on their cellular source and cargo. Exosomes secreted by antigen-presenting cells can attenuate or stimulate immune responses, carrying functional MHC-peptide complexes that modulate T cell responses. They can activate T and B cells, modulate macrophage function, and influence natural killer (NK) cell activity.[^5][^19][^20]

MSC-derived exosomes possess immunomodulatory and anti-inflammatory properties that can reduce chronic inflammation, inhibit inflammatory factors, and promote the production of anti-inflammatory cytokines like IL-10. This makes them particularly valuable for treating autoimmune conditions and inflammatory disorders.[^21][^1][^2][^6][^5]

- - Wound Healing and Tissue Repair

Exosomes regulate all phases of skin wound healing—inflammation, proliferation, and remodeling—primarily through the actions of circulating microRNAs. They facilitate wound closure by reducing inflammation, promoting cell proliferation and migration (especially of fibroblasts and endothelial cells), boosting angiogenesis, and regulating extracellular matrix remodeling.[^22][^23][^24][^25]

Studies show that exosome administration significantly improves wound closure rates and increases vascularity in wound sites. MSC-derived exosomes can reverse acute thermal injury-induced cell apoptosis, accelerate healing, and are particularly effective in treating chronic and diabetic wounds.[^23][^24][^26]

- - Cancer Treatment and Drug Delivery

Exosomes are being developed as sophisticated drug delivery vehicles for cancer therapy, offering several advantages over traditional nanocarriers. Their lipid bilayer membranes can encapsulate and safely deliver various therapeutic cargoes including chemotherapeutic drugs (doxorubicin, paclitaxel), nucleic acids (siRNA, miRNA), and proteins directly to cancer cells. Exosomes demonstrate high biocompatibility, low immunogenicity, extended drug half-life, and the ability to overcome multidrug resistance in cancer cells.[^17][^27][^28][^29]

Engineered exosomes can be modified with targeting molecules to achieve tissue-specific delivery, increasing drug accumulation at tumor sites while reducing systemic toxicity. They can also pass through the blood-brain barrier, making them valuable for treating brain tumors.[^27][^28][^17]

- - Metabolic Health and Diabetes Management

Exosomes show therapeutic potential in treating both Type 1 and Type 2 diabetes mellitus by modulating immune responses, reducing insulin resistance, promoting pancreatic beta-cell regeneration, and restoring insulin-secreting function. MSC-derived exosomes can decrease blood glucose levels, increase plasma insulin, enhance glucose metabolism in the liver, and improve glucose transporter (GLUT4) function in muscle tissue.[^30][^31][^32]

In Type 1 diabetes, exosomes reduce pro-inflammatory cytokines and increase regulatory T cell populations. For Type 2 diabetes, they can reverse insulin resistance, enhance glycogen storage, suppress beta-cell apoptosis, and stimulate islet regeneration.[^31][^30]

- - Anti-Aging and Skin Rejuvenation

Exosomes combat cellular aging by stimulating collagen and elastin production, improving skin elasticity, reducing wrinkles, and promoting tissue regeneration. They work at the cellular level to boost skin cell regeneration, enhance blood circulation, and counteract the decreased cell turnover that accelerates aging. Stem cell-derived exosomes can restore skin physiological function through decreased expression of matrix metalloproteinases, increased collagen production, and modulation of intracellular signaling pathways.[^33][^34][^35][^36][^6]

Exosome therapy is increasingly used in aesthetic medicine, often combined with microneedling or other procedures to accelerate results and improve the appearance of sun damage, aging, pigmentation, and rosacea.[^34][^37][^38][^39]

- - Disease Diagnosis and Biomarkers

Beyond their therapeutic applications, exosomes serve as valuable diagnostic biomarkers for numerous diseases including cancer, cardiovascular disease, neurological disorders, and metabolic conditions. Because exosomes circulate in bodily fluids like blood, urine, and cerebrospinal fluid, they enable non-invasive "liquid biopsies" for real-time disease monitoring and early detection.[^40][^41][^42][^2]

Cancer-derived exosomes contain molecular signatures reflecting genetic and phenotypic alterations in their cells of origin, making them useful for diagnosing various cancers, predicting treatment responses, and monitoring disease progression. Exosomal PD-L1, for example, serves as a potential predictor of response to anti-PD-1 immunotherapy in melanoma and lung cancer.[^41]

- - Clinical Safety and Emerging Applications

Clinical trials have demonstrated that exosome-based therapies are generally safe with minimal adverse events. A systematic review and meta-analysis found that extracellular vesicle-based therapy showed a low incidence of serious adverse events (0.7%) and overall adverse events (4.4%). Patients receiving MSC-derived exosomes have tolerated treatments well without evidence of acute toxicity, immune reactions, or clinical instability.[^43][^44][^45][^46]

Current clinical trials are investigating exosome therapies for COVID-19, Alzheimer's disease, inflammatory bowel disease, and various other conditions, with early results showing promise for safety and efficacy. However, challenges remain in standardizing exosome isolation, purification, characterization methods, and scaling up production for widespread clinical application.[^4][^45][^47][^46][^2][^43]

Exosomes represent a transformative approach to medicine, offering cell-free therapeutic alternatives that harness the body's natural healing mechanisms while avoiding many risks associated with cell-based therapies. Their multifaceted roles in intercellular communication, tissue regeneration, immune modulation, and drug delivery position them as powerful tools for treating a wide spectrum of human diseases and supporting overall health.[^48][^1][^3] [^49][^50][^51][^52][^53][^54][^55][^56][^57][^58][^59][^60][^61][^62][^63][^64][^65][^66][^67][^68][^69][^70][^71][^72][^73][^74][^75][^76][^77]


The History of Exosomes

The discovery and understanding of exosomes has evolved dramatically over several decades, transforming from observations of mysterious cellular debris into recognition of these vesicles as sophisticated biological messengers with immense therapeutic potential.

- - Early Observations: The "Platelet Dust" Era (1940s-1960s)

The first glimpse of what would later be recognized as extracellular vesicles came in 1946, when scientists Chargaff and West identified particulate fractions with coagulant activity while isolating blood-coagulating proteins from plasma using high-speed centrifugation. This provided the initial hint that vesicular structures existed in biological fluids, though their nature remained unclear.[^1]

Two decades later in 1967, Peter Wolf at the University of Birmingham made a landmark observation while studying blood coagulation. Using ultracentrifugation and electron microscopy, Wolf visualized platelet-derived particulate structures of varying diameter and density in blood samples, which he termed "platelet dust". He demonstrated that this material was sedimentable by high-speed centrifugation, rich in phospholipids, originated from platelets, and possessed coagulation properties identical to Platelet Factor 3. Wolf's work established that these particles were present in both plasma and serum and played an active role in the coagulation process.[^2][^3][^4][^1]

- - Defining Extracellular Vesicles (1970s-1980s)

The term "exosome" itself has an interesting etymological history. It was first introduced in the 1970s, though initially referring to DNA fragments transferred between cells rather than the membrane-bound vesicles we recognize today. In 1971, Aaronson and colleagues were the first to use the term "extracellular vesicles" in scientific literature.[^5][^6]

In 1981, Trams and colleagues proposed using the term "exosome" to describe the exfoliation of microvesicles from the plasma membrane. Their work revealed that the exfoliation of membranous vesicles occurred in many different normal and neoplastic cells, and that this exfoliative process was selective, suggesting these vesicles consisted of specific domains of the plasma membrane.[^5]

- - The Johnstone Discovery: Birth of Modern Exosome Research (1983)

The pivotal breakthrough that established modern exosome research came simultaneously in 1983 when two independent research groups—one led by Rose Johnstone at McGill University and another by Philip Stahl—published complementary papers describing extracellular vesicles. These studies investigated the maturation of reticulocytes (immature red blood cells) into erythrocytes and the loss of transferrin receptors from the cell surface.[^7][^8][^9][^10][^11]

Harding, Heuser, and Stahl demonstrated for the first time that late endosomes called "multivesicular endosomes" (MVEs) were responsible for recycling the transferrin receptor. To their surprise, they found that MVEs fused with the plasma membrane, leading to the release of small vesicles under 100 nanometers in diameter that were found within the MVEs. Their electron microscopy images provided direct visual evidence of this MVE externalization event.[^9][^11]

Simultaneously, Pan and Johnstone proved in sheep reticulocytes that the transferrin receptor was externalized via vesicles. Although they did not directly visualize the phenomenon by electron microscopy, they used anti-transferrin-receptor antibodies to follow the trafficking of the protein. Pan and Johnstone initially favored a mechanism of vesicle shedding at the plasma membrane.[^11][^7][^9]

Rose Johnstone formally coined the term "exosome" in 1987 to describe these secreted vesicles. Born Rose Mamelak in Poland in 1928, Johnstone arrived in Montreal in 1936 and went on to become the first woman to chair McGill University's Department of Biochemistry. Her discovery of exosomes and their unique secretion pathway distinguished them from other types of vesicles. In a 2005 retrospective paper titled "Revisiting the road to the discovery of exosomes," Johnstone described the long, uncertain process of trial and error that led to identifying "these small structures, of relatively modest protein content".[^8][^12][^10][^13][^14]

- - The Dormant Period (1987-1996)

Following these initial discoveries, exosomes did not immediately capture widespread scientific attention. Initially thought to function merely as a cellular waste disposal system—a kind of "cellular trash can"—exosomes were not recognized for their true biological significance. Research publications on exosomes remained limited during this period, with fewer than 10 publications annually before 2004.[^12][^15][^16][^17][^7]

- - The Immunological Breakthrough (1996)

The field experienced its first major breakthrough in 1996 when Raposo and colleagues demonstrated that B lymphocytes secrete exosomes containing MHC class II molecules from a specialized late endocytic compartment called MIIC (MHC class II-enriched compartment). Critically, these secreted exosomes could induce antigen-specific MHC class II-restricted T cell responses. This landmark study revealed that exosomes were not cellular waste but rather functional mediators of immune communication.[^15][^18][^19][^6][^11]

Raposo's work showed that the limiting membrane of MIICs could fuse directly with the plasma membrane, resulting in release of internal MHC class II-containing vesicles. The overall surface protein composition of exosomes differed significantly from that of the plasma membrane, and exosome-bound MHC class II was in a compact, peptide-bound conformation capable of presenting antigens to T cells. This discovery opened researchers' eyes to the functional importance of exosomes in immunology and intercellular communication.[^18][^20][^19][^11][^15]

- - The RNA Revolution (2007)

The second major breakthrough in exosome research came in 2007 when Hadi Valadi, Jan Lötvall, and colleagues at the University of Gothenburg, Sweden, made a groundbreaking discovery: exosomes contain both mRNA and microRNA that can be transferred between cells and remain functional in recipient cells. This finding opened an entirely new field of inquiry into genetic exchange between cells.[^21][^22][^23][^8][^11][^15]

Their research showed that exosomes from mouse and human mast cells contained mRNA from approximately 1,300 genes, many of which were not present in the cytoplasm of the donor cell. In vitro translation proved that the exosomal mRNAs were functional. After transfer of mouse exosomal RNA to human mast cells, new mouse proteins were found in the recipient cells, demonstrating that transferred exosomal mRNA could be translated after entering another cell. Valadi and colleagues proposed calling this transferred material "exosomal shuttle RNA" (esRNA).[^22][^23]

This discovery transformed understanding of exosomes from simple protein carriers to sophisticated vehicles capable of transferring genetic information and reprogramming recipient cells. Initial skepticism about RNA presence in exosomes being incidental was subsequently ruled out by demonstrations showing specificity in exosomal RNA cargo.[^11][^21]

Exponential Growth and Institutionalization (2000s-2010s)

Following the RNA discovery, exosome research entered a phase of explosive growth. Research publications began steadily increasing from 2005 to 2013, followed by a surge from 2014 to 2020 that achieved a breakthrough from triple to quadruple digits annually. By 2021, the field reached its heyday with more than 1,000 publications each year.[^24][^17][^7][^11]

Over the past 11 years leading up to 2022, the number of exosome-related publications on PubMed rose from 282 to 5,073, representing a 30% compound annual growth rate. As of December 2024, over 8,000 publications specifically on exosomes in cancer research alone had been identified. By 2025, researchers had published more than 8,400 papers on exosomes since Johnstone's discovery.[^17][^25][^12]

Recognizing the need for standardization and collaboration in this rapidly expanding field, the International Society for Extracellular Vesicles (ISEV) was established in 2011. Founded with Jan Lötvall as its first president, ISEV became a nonprofit organization dedicated to advancing global extracellular vesicle research. The society's formation was a crucial step in creating uniformity, sharing research ideas, and providing guidance for EV research during a period when there was little rigor and standardization.[^26][^27][^6][^28]

ISEV has published influential consensus guidelines including the "Minimal Information for Studies of EVs" (MISEV), with versions released in 2014 (MISEV2014), 2018 (MISEV2018), and 2023 (MISEV2023) to improve rigor and standardization in EV studies across different research groups. The society publishes two journals—the Journal of Extracellular Vesicles and the Journal of Extracellular Biology —and hosts annual meetings that bring together the field's leading researchers. With nearly 2,000 members by 2025, ISEV has become the largest community of extracellular vesicle researchers in the world.[^29][^27][^30][^26]

- - Nomenclature Evolution

The terminology surrounding exosomes has evolved considerably and remains somewhat complex. Initially, researchers stratified extracellular vesicles based on size: exosomes (<100 nm), microvesicles or microparticles (100-1,000 nm), and apoptotic bodies (>1,000 nm). However, this size-based classification proved problematic because isolation methods often could not reliably distinguish between different vesicle types based solely on physical properties.[^3][^6][^24][^5]

From the late 1990s to early 2010s, researchers used terms like "exosomes," "microvesicles," "ectosomes," "membrane particles," "exosome-like vesicles," and "apoptotic vesicles" somewhat interchangeably. The term "exosome" has been used in at least three different ways: following the original biogenetic definition (vesicles that bud into endosomes and are released when MVBs fuse with the plasma membrane), using the original broad definition by Trams, or as a catch-all term for small extracellular vesicles.[^5]

Following the MISEV2018 guidelines, the field has moved toward using operational terms that can be experimentally verified, with "extracellular vesicles" (EVs) serving as an all-encompassing term when the precise origin of isolated vesicles cannot be ascertained. This nomenclature shift reflects the practical challenges of distinguishing vesicle subtypes while maintaining scientific rigor.[^27][^6][^31][^5]

- - Therapeutic Development and Clinical Translation (2010s-2020s)

The 2010s witnessed a paradigm shift toward therapeutic applications of exosomes. At Mayo Clinic, researchers Atta Behfar and Andre Terzic pioneered work translating exosome research into clinical products. Initially focused on stem cell therapies for cardiac regeneration in the early 2000s, by 2010 they discovered that stem cells exerted their therapeutic effects primarily through exosomes they secreted rather than through direct cellular action.[^32][^33][^34]

A major challenge was that stem cells were expensive and difficult to scale for commercial production. Behfar and Terzic made a breakthrough by discovering they could use platelets—abundant blood cells—as the source for therapeutic exosomes instead of stem cells. They engineered platelet-derived exosomes containing specific regenerative molecules, creating a purified product that could sit on a shelf at room temperature for a year while maintaining potency. This work, commercialized through Mayo's Employee Entrepreneurship Program, represented a major step toward making exosome therapies practical and scalable.[^33][^32]

By the mid-2010s, researchers began engineering exosomes to enhance their therapeutic capabilities. Modification strategies included loading exosomes with drugs, nucleic acids, or proteins (internal modification) and altering their surface properties for targeted delivery (external modification). These engineered exosomes demonstrated potential for treating inflammatory diseases, cancer, neurological disorders, and numerous other conditions.[^34][^35][^36][^37]

- - Market Growth and Commercialization (2020s)

The exosome research and therapeutic market has experienced remarkable growth. The global exosome diagnostics and therapeutics market was valued at just \$16.1 million in 2016, grew to \$97.8 million in 2021, reached \$227.5 million in 2023, stood at \$189.4 million (for research products) to \$214.4 million in 2025, and is projected to reach \$480.6-512.6 million by 2026-2028, and potentially \$1.3 billion by 2028. This represents compound annual growth rates ranging from 17.5% to 47.3% depending on the market segment.[^38][^39]

As of 2024, over 70 major companies are actively developing exosome-based therapies, with more than 80 pipeline therapies in various stages of clinical development. At least 150 clinical trials registered on ClinicalTrials.gov are investigating exosome-based therapies for various diseases, with 31 trials specifically exploring mesenchymal stem cell-derived exosomes. The majority of therapeutic applications focus on cancer and neurodegenerative diseases.[^40][^41][^17]

- - Current State and Future Directions

Today, exosomes are recognized as sophisticated biological messengers with roles spanning developmental biology, immunity, tissue regeneration, disease pathology, and drug delivery. They have graduated from "background noise to headline acts in oncology, immunology, vaccines, and regenerative medicine".[^42][^35][^7][^40]

Despite this progress, significant challenges remain in manufacturing, quality control, standardization, and regulatory approval. Production yields remain low, purification is challenging, and regulatory standards are still evolving. The role of specialized Contract Development and Manufacturing Organizations (CDMOs) focused on exosome production has become essential for translating laboratory discoveries into scalable, GMP-compliant therapies.[^35][^37][^40][^34][^42]

Rose Johnstone, who passed away in 2009, could scarcely have predicted that her serendipitous discovery while studying red blood cells would enable the development of entirely new kinds of medicines with potential to transform treatment of a wide range of diseases. As she herself wrote in 2003, "In science, predicting the significance and outcome of really new work eludes many of us". Her "Alice in Blunderland approach" to discovering exosomes has opened pathways to what may become one of the most transformative therapeutic platforms of the 21st century.[^10][^14][^12][^24] [^43][^44][^45][^46][^47][^48][^49][^50][^51][^52][^53][^54][^55][^56][^57][^58][^59][^60][^61][^62][^63][^64][^65][^66][^67][^68][^69][^70][^71][^72][^73][^74]

Click here for all Perplexity references


The Hidden Language of Life: How Exosomes Carry Nature’s Instructions to Your Cells

- - Sayer Ji (67 minute video, Nov 2025)


See related in VitaminDWiki

*Microbiome

*Gut microbiome massively changed by weekly vitamin D – July 2015

*Nutrients, Microbiome, and Chronic Diseases - April 2023

*Gut microbiome altered by many nutrients – such as Vitamin D