Fact-checked by Grok 2 weeks ago

Plasma cell

A plasma cell is a terminally differentiated B lymphocyte, a type of , that serves as the primary producer of antibodies in the humoral , secreting thousands of immunoglobulin molecules per second to combat pathogens and maintain long-term immunity. These cells arise from activated B cells following antigenic stimulation, undergoing a maturation process driven by transcription factors such as Blimp-1, , and XBP-1, which reprogram the cell to prioritize antibody synthesis over proliferation. Morphologically, plasma cells are larger than typical s (14–20 micrometers in diameter), featuring an eccentric nucleus with a characteristic "clock-face" pattern, abundant rough for protein production, and a prominent perinuclear Golgi apparatus, often marked by surface expression of and CD138. Beyond antibody secretion, plasma cells exhibit diverse functions, including the production of immunomodulatory cytokines like IL-10, IL-35, and IL-17, which regulate , inflammation, and even hematopoiesis in various tissues. Long-lived plasma cells, capable of persisting for decades in survival niches such as the and gut mucosa, ensure sustained serological against infections and vaccines, with the majority residing in niches supported by stromal cells, while others persist in mucosal tissues like the gut. Dysregulation of plasma cells contributes to immunodeficiencies (e.g., agammaglobulinemia from impaired differentiation) and pathologies like or autoimmune diseases, where autoreactive plasma cells drive chronic antibody-mediated damage. Plasma cells were first described in by Wilhelm Waldeyer, with their role as the primary antibody-producing cells confirmed experimentally in the mid-20th century; they remain central to understanding adaptive immunity and therapeutic targeting in disorders.

Structure

Morphology

Plasma cells are ovoid or round cells typically measuring 9 to 20 μm in diameter, with a mean of approximately 14 μm, making them larger than naive B cells which are around 7-10 μm. Under light microscopy, they exhibit a characteristic basophilic cytoplasm due to the abundance of rough endoplasmic reticulum (RER) involved in protein synthesis, an eccentric nucleus, and a perinuclear halo corresponding to the Golgi apparatus region. The nucleus is round or oval, positioned off-center, and features coarse heterochromatin arranged in a distinctive clock-face or cartwheel pattern, with clumps of chromatin distributed along the nuclear periphery. Electron microscopy reveals further ultrastructural details that underscore their specialization for antibody production. The cytoplasm is dominated by extensive arrays of RER, appearing as parallel stacks of flattened cisternae studded with ribosomes, often containing granular material within the lumen. A prominent Golgi apparatus occupies the perinuclear halo, consisting of vesicles, tubules, and dense globules for processing and packaging proteins. The nucleus shows a higher nucleo-cytoplasmic ratio in immature forms with well-developed nucleoli, while mature plasma cells have a denser, eccentric nucleus with less prominent nucleoli. Occasionally, cytoplasmic inclusions such as Russell bodies—dilated RER sacs filled with dense material—may be observed, reflecting accumulated immunoglobulins.

Surface antigens

Plasma cells are distinguished from other immune cells by a unique profile of surface antigens that reflect their terminal differentiation from B lymphocytes. The primary identifier is CD138 (also known as syndecan-1), a highly expressed on the surface of mature plasma cells, which facilitates their adhesion to components in survival niches. Co-expression of , a multifunctional ectoenzyme, at high levels further characterizes plasma cells, enabling their robust detection in both normal and pathological contexts. In contrast, B cell-associated markers such as and are downregulated or absent; CD19 expression is low or negative on mature plasma cells, while CD20 is typically not expressed, marking the loss of identity. During differentiation from activated B cells, plasma cells undergo significant changes in surface antigen expression, including the loss of major histocompatibility complex class II (MHC II) molecules and the . MHC II, which is highly expressed on B cells for , is progressively downregulated as plasmablasts mature into long-lived plasma cells, correlating with their reduced role in T cell interactions. Similarly, surface BCR (membrane-bound immunoglobulin) is lost or markedly reduced, shifting the cell's focus from recognition to antibody secretion; this is particularly evident in IgG-producing plasma cells, though some IgA- and IgM-secreting cells may retain low levels. These alterations begin in plasmablasts, which often retain partial B cell marker expression (e.g., CD19 positive, CD138 low), and culminate in mature plasma cells with a streamlined profile dominated by CD138 and CD38. Adhesion molecules like , a hyaluronan receptor, are upregulated on plasma cells to support their homing to niches, where interactions with stromal cells promote survival. This homing is critical for long-lived plasma cells, which express high CD44 levels to facilitate retention in supportive microenvironments. In diagnostics, these surface antigens are essential for identifying plasma cells via and . panels typically gate on CD38high CD138+ populations, with absence of and confirming maturity and distinguishing normal from neoplastic cells, such as in where aberrant expression (e.g., CD19- CD56+) aids in detecting . relies heavily on CD138 for visualizing plasma cells in tissue biopsies, offering high specificity for plasmacytic infiltrates. These markers' expression patterns also correlate with morphological features, such as the eccentric in CD138+ cells observed in aspirates.

Development and Lifespan

Differentiation from B cells

Plasma cells differentiate from mature B cells through activation pathways that initiate the transition to antibody-secreting cells. In T cell-dependent activation, which occurs primarily in germinal centers of secondary lymphoid organs, naive B cells recognize antigens via their (BCR) and receive co-stimulatory signals from CD4+ helper T cells, leading to proliferation and differentiation into plasmablasts. T cell-independent activation, in contrast, involves direct stimulation of B cells by antigens with repetitive structures, such as , bypassing T cell help and resulting in more rapid but lower-affinity responses, often producing IgM-secreting plasmablasts. Key signals driving this differentiation include CD40 ligand (CD40L) expressed on activated T cells, which binds CD40 on to promote survival and proliferation, synergizing with cytokines such as interleukin-4 (IL-4) and IL-21. IL-4 supports growth and class switching to IgG and IgE, while IL-21 potently induces plasmablast formation and immunoglobulin secretion by upregulating terminal differentiation programs. Additionally, B cell-activating factor (BAFF, also known as BLyS) and a proliferation-inducing ligand () provide survival signals through receptors like TACI and BCMA, enhancing the commitment to the plasma cell lineage. The transcriptional program for plasma cell differentiation is orchestrated by key factors, including and (encoding Blimp-1). initiates the process by activating expression and repressing identity genes, while Blimp-1 further enforces the plasmacytic state by silencing proliferation-associated genes like and promoting immunoglobulin secretion machinery. These factors act in a dose-dependent manner, with graded levels coordinating transitions from activated to plasmablasts. Plasmablasts emerge as the immediate precursors to plasma cells, characterized by initial secretion of immunoglobulins, predominantly IgM in early responses. In germinal centers during T cell-dependent responses, B cells undergo affinity maturation through , which introduces mutations in antibody variable regions to enhance binding, followed by selection of high-affinity clones. Class-switch recombination also occurs here, diversifying isotypes from IgM to IgG, IgA, or IgE under the influence of cytokines like IL-4 or IFN-γ, preparing plasmablasts for effector functions. This differentiation process unfolds rapidly following exposure, with activated B cells proliferating and forming plasmablasts within 3–7 days , depending on the pathway and type.

Immature and short-lived plasma cells

Immature and short-lived plasma cells arise as proliferating antibody-secreting cells primarily in extrafollicular regions of secondary lymphoid organs, such as the red pulp of the and medullary cords of lymph nodes, where they expand rapidly in response to antigenic stimulation. These cells transition from plasmablasts, the immediate proliferating precursors derived from activated B cells under influences like IL-21, and are characterized by markers such as expression in humans and B220 in mice, reflecting their immature state. Their high proliferative capacity enables a swift buildup during the early phase of an , generating large numbers—often in the millions within the —to mount an immediate defense. These plasma cells exhibit a brief lifespan of approximately 3 to 5 days, constrained by limited access to survival signals in their transient niches within lymphoid tissues. Unlike more persistent counterparts, they lack robust support from stromal cells or cytokines, leading to rapid turnover and contributing to the resolution of acute responses. In terms of function, they predominantly secrete low- IgM antibodies, providing essential but non-specialized humoral support to control early dissemination before affinity maturation occurs. Apoptosis in these cells is tightly regulated by the of proteins, where insufficient anti-apoptotic members like result in heightened sensitivity to intrinsic death pathways triggered by bioenergetic stress or overload from high secretory demands. They also display vulnerability to Fas-mediated in precursor stages, facilitating the elimination of excess cells post-initial response to prevent overproduction. Overall, while comprising the majority of antibody-secreting cells in the acute phase, most undergo programmed death shortly after, ensuring a controlled and temporary contribution to immunity.

Long-lived plasma cells and survival niches

Following differentiation in germinal centers, a subset of plasma cells acquires competence for homing to the through upregulation of the CXCR4, which interacts with its ligand SDF-1 () produced by bone marrow stromal cells, directing migration and retention in survival niches. This /SDF-1 axis is essential for post-germinal center plasma cells to exit peripheral sites and establish residence in the , where they compete for limited niche space. Within the , long-lived plasma cells occupy specialized survival niches formed by interactions with stromal cells, including mesenchymal progenitors and perivascular reticular cells, which provide essential survival signals. Key factors in these niches include and BAFF, secreted by stromal cells and non-hematopoietic nurse-like cells, which bind to receptors such as and TACI on plasma cells to promote through anti-apoptotic pathways. Additionally, IL-6 signaling from niche-associated cells enhances plasma cell persistence by supporting metabolic fitness and inhibiting cell death. These extrinsic cues enable a small population of plasma cells, comprising approximately 1% of total bone marrow cells, to maintain high output despite their scarcity. Long-lived plasma cells enter a non-proliferative, quiescent state characterized by repression of the transcription factor Bach2, which facilitates terminal and suppresses proliferation genes, ensuring long-term survival without division. Elevated expression of on these cells further contributes to quiescence by mediating adhesion to components in the niche, stabilizing their position and reducing motility. This quiescent allows plasma cells to persist for decades, as evidenced by sustained tetanus-specific immunity in humans for over 10 years post-vaccination, underpinning serological memory. Recent research highlights the role of hypoxia-inducible factors (HIFs), particularly HIF-1α, in maintaining these niches under the bone marrow's low-oxygen environment (PO₂ ~2-3%), where it upregulates stromal-derived factors like to support plasma cell adhesion and survival. Metabolic adaptations driven by HIF signaling, including enhanced and mitochondrial function, enable long-lived plasma cells to meet the energy demands of continuous secretion while resisting , further promoting their longevity.

Function

Antibody secretion

Plasma cells are highly specialized for the high-rate secretion of , producing up to 10,000 immunoglobulin molecules per cell per second through an expanded () and Golgi apparatus pathway. This secretory capacity arises from their morphological adaptations, including abundant , which supports the massive protein synthesis required for antibody production. The process begins with the of immunoglobulin heavy and light chains on polysomes associated with the membrane, followed by their translocation into the lumen for folding and assembly. Within the ER, heavy and light chains undergo proper folding, assisted by chaperones, and form disulfide bonds to assemble into complete immunoglobulin monomers or multimers, with N-linked glycosylation occurring to stabilize the structure and facilitate trafficking. Glycosylation patterns vary by isotype, influencing antibody function and stability during secretion. Plasma cells produce diverse isotypes—IgG, IgA, IgE, and IgM—determined by prior class-switch recombination in B cell precursors, allowing adaptation to different immune challenges. The transcription factor XBP1 plays a central role in expanding the secretory apparatus by upregulating genes for ER biogenesis, protein folding, and vesicular transport, enabling sustained high-output antibody production. To manage the resulting ER stress from this intense synthetic load, plasma cells activate a specialized unfolded protein response (UPR) that selectively engages the IRE1-XBP1 arm while suppressing the PERK branch, thereby promoting survival and secretion without triggering apoptosis. Secretion rates can be quantified ex vivo using enzyme-linked immunospot (ELISPOT) assays, which detect antibody spots formed by individual cells, revealing heterogeneity in output among plasma cell populations.

Role in humoral immunity

Plasma cells serve as the primary effectors of the by secreting antibodies that neutralize and facilitate opsonization, thereby marking infectious agents for by immune cells such as macrophages and neutrophils. These antibodies bind to specific epitopes on , preventing their attachment to host cells and inhibiting replication, while opsonization enhances the efficiency of pathogen clearance through Fc receptor-mediated uptake. In the context of immunological memory, long-lived plasma cells provide sustained by continuously producing long after the initial encounter, operating independently of T cell support following their differentiation from B cells. This autonomy ensures persistent protection against reinfection without the need for ongoing antigenic stimulation or T cell activation. Different isotypes produced by plasma cells contribute to site-specific defense mechanisms; for instance, IgG antibodies dominate systemic circulation, providing broad protection against blood-borne pathogens, whereas IgA antibodies are secreted at mucosal surfaces to reinforce barriers in the respiratory, gastrointestinal, and urogenital tracts. Once migrated to survival niches such as the or mucosal tissues, plasma cells exhibit minimal direct interactions with other immune cells, instead delivering protection primarily through the release of soluble into the bloodstream or local secretions. Beyond antibody production, plasma cells can secrete immunomodulatory cytokines such as interleukin-10 (IL-10), IL-35, and IL-17, which contribute to regulatory functions in immunity. IL-10 and IL-35 from regulatory plasma cells promote and suppress excessive , while IL-17 production by certain plasma cell subsets may enhance responses against extracellular pathogens or contribute to autoimmune pathology. The functional role of plasma cells in is evolutionarily conserved across vertebrates, where B cell-derived plasma cell-like effectors generate adaptive responses to combat diverse pathogens. Quantitatively, these cells maintain lifelong levels, as exemplified by measles-specific IgG titers with an estimated half-life exceeding 3,000 years, ensuring durable protection against reinfection.

Clinical Significance

Plasma cell neoplasms and disorders

Plasma cell neoplasms represent a group of disorders characterized by the uncontrolled of clonal plasma cells, leading to excessive production of monoclonal immunoglobulins or chains. , the most common plasma cell , involves the clonal expansion of malignant plasma cells in the , often exceeding 10% of cellularity, which disrupts normal hematopoiesis and . This results in the classic CRAB symptoms: hypercalcemia due to osteolytic , renal failure from chain cast nephropathy or hypercalcemia, from infiltration and cytokine-mediated suppression, and bone lesions manifesting as lytic lesions, fractures, or . Related premalignant and malignant disorders include (MGUS), , and . MGUS is an asymptomatic precursor condition defined by a serum monoclonal protein less than 3 g/dL, clonal plasma cells less than 10%, and absence of end-organ damage or myeloma-defining events. features an IgM monoclonal protein produced by lymphoplasmacytic cells in the , often causing , , and due to IgM-mediated effects. In , misfolded light chains secreted by clonal cells deposit as amyloid fibrils in organs, particularly the heart and kidneys, leading to and . Dysfunction or overactivity of plasma cells also contributes to autoimmune diseases through the persistent production of pathogenic autoantibodies. In systemic lupus erythematosus (SLE), autoreactive plasma cells secrete antibodies such as anti-double-stranded DNA and anti-Smith, driving immune complex deposition and tissue in organs like the kidneys and skin. Similarly, in , long-lived plasma cells in the synovium produce and anti-citrullinated protein antibodies, perpetuating and via autoantibody-mediated complement and immune . Plasma cell deficiencies underlie certain immunodeficiencies, resulting in impaired antibody production and recurrent infections. (CVID) involves defective B-cell differentiation into plasma cells, leading to , reduced switched memory B cells, and paucity of plasma cells in tissues, which predisposes to sinopulmonary infections and . Agammaglobulinemia, often X-linked, features an absence of mature B cells and thus plasma cells due to mutations in the gene, causing profound and susceptibility to bacterial infections from early infancy. Diagnosis of plasma cell neoplasms typically relies on laboratory and histopathological findings. identifies monoclonal proteins (M-spikes) in over 80% of cases, while confirms the immunoglobulin type; urine protein electrophoresis detects light chains (Bence Jones proteins). is essential, revealing clonal plasma cells often positive for CD138, with a of 10% or greater (≥10%) clonal cells supporting a diagnosis of when combined with myeloma-defining events. Post-2020 research has identified prolonged production, such as antinuclear and antiphospholipid antibodies, in survivors, contributing to symptoms like and neurological dysfunction. This immune dysregulation may stem from SARS-CoV-2-induced B-cell hyperactivity, with new-onset autoantibodies persisting beyond 12 months in a subset of patients.

Therapeutic targeting and immunomodulation

Proteasome inhibitors, such as , target the unfolded protein response (UPR) in plasma cells, which are highly sensitive due to their intense immunoglobulin synthesis and proteasomal workload. In , induces by overwhelming the proteasome's capacity to degrade misfolded proteins accumulated during UPR activation. This mechanism has established as a cornerstone therapy, improving response rates and survival in myeloma patients. Monoclonal antibodies like daratumumab target CD38, a surface antigen highly expressed on malignant plasma cells, inducing cell death through complement-dependent cytotoxicity, antibody-dependent cellular cytotoxicity, and phagocytosis. Approved for relapsed or refractory multiple myeloma, daratumumab monotherapy yields objective response rates of around 30% in heavily pretreated patients, with enhanced efficacy in combinations. Elotuzumab, targeting SLAMF7 (CD319), promotes natural killer cell-mediated antibody-dependent cellular cytotoxicity against myeloma cells while directly activating NK cells. Expressed uniformly on plasma cells, SLAMF7 enables elotuzumab to achieve progression-free survival benefits when combined with lenalidomide and dexamethasone in relapsed myeloma. Immunomodulatory drugs like lenalidomide enhance immune surveillance by activating T cells and natural killer cells, facilitating clearance of malignant plasma cells in multiple myeloma. Lenalidomide reduces programmed death-1 expression on T and NK cells, boosts interleukin-2 and interferon-γ secretion, and degrades the transcription factor Ikaros to promote cytotoxic responses. This leads to improved overall survival in combination regimens for both newly diagnosed and relapsed disease. Post-2020 emerging therapies include BCMA-targeted chimeric antigen receptor T-cell (CAR-T) therapy, such as idecabtagene vicleucel, approved in 2021 for relapsed or refractory after at least four prior lines of therapy. Targeting (BCMA) on cells, idecabtagene vicleucel achieves deep responses, with 73% overall response rates and 33% complete responses in pivotal trials. Bispecific antibodies, such as (BCMA-CD3) and (GPRC5D-CD3), redirect T cells to lyse malignant cells, yielding response rates exceeding 60% in heavily pretreated patients. Recent 2025 data from real-world studies and combinations (e.g., with ) confirm response rates exceeding 70-80% in relapsed/refractory cases. Inhibitors of APRIL and BAFF disrupt survival niches in the by blocking these cytokines essential for long-lived cell maintenance, leading to rapid depletion of cells in preclinical models. In , B-cell depletion with rituximab indirectly reduces plasma cell precursors by targeting CD20-positive B cells, limiting replenishment of autoantibody-secreting plasma cells. This approach depletes short-lived autoreactive plasma cells in diseases like and , though long-lived plasma cells persist. Plasma exchange removes circulating pathogenic antibodies produced by plasma cells, providing rapid symptom relief in antibody-mediated autoimmune conditions such as Guillain-Barré syndrome and . By filtering and replacing it with or donor , this procedure reduces autoantibody levels by 50-70% per session. mRNA vaccines, such as those for SARS-CoV-2, boost the generation of long-lived plasma cells in the bone marrow to enhance humoral memory and sustained antibody production. These vaccines induce robust germinal center responses, leading to antigen-specific memory B cells that differentiate into long-lived plasma cells secreting neutralizing antibodies for months to years post-vaccination.

References

  1. [1]
    Histology, Plasma Cells - StatPearls - NCBI Bookshelf - NIH
    Plasma cells are differentiated B-lymphocyte white blood cells capable of secreting immunoglobulin or antibodies. They play a significant role in the adaptive ...
  2. [2]
  3. [3]
    The Structure of Lymphocytes and Plasma Cells | Oncohema Key
    Jun 14, 2016 · The mature plasma cell has a characteristic basophilic cytoplasm and an eccentric nucleus when treated with a polychrome stain. The nuclear ...
  4. [4]
    Distribution, location, and ultrastructure of plasma cells in the ...
    Jun 1, 2009 · Parenchymal plasma cells exhibited typical ultrastructure with abundant, flattened stacks of rough endoplasmic reticulum, and hypertrophied ...
  5. [5]
    The fine structure of connective tissue: II. The plasma cell
    When observed with the light microscope, mature plasma cells have a basophilic cytoplasm, a perinuclear clear area, and an eccentric dense nucleus.
  6. [6]
    Immunophenotyping in multiple myeloma and related plasma cell ...
    Role of flow cytometry in the diagnosis and classification of plasma cell diseases ... Gating on CD38 and CD138 positive plasma cells reveals both CD45 ...Missing: MHC | Show results with:MHC
  7. [7]
    The Maintenance of Memory Plasma Cells - PMC - PubMed Central
    Human plasma cells are characterized by the co-expression of CD138 and CD38, which allows for the identification of plasma cells in the bone marrow or in cell ...
  8. [8]
    Targeting B Cells and Plasma Cells in Autoimmune Diseases
    In this review, we will discuss the multiple pro- and anti-inflammatory roles of B lineage cells play in autoimmune diseases.Missing: loss CD44
  9. [9]
    Progressive differentiation toward the long-lived plasma cell ...
    Dec 14, 2022 · Antibodies (Abs) are produced by terminally differentiated plasma cells (PCs) and have distinct functions depending on their isotype.
  10. [10]
    Circulating human B and plasma cells. Age-associated changes in ...
    With age, there is an inverse correlation with the percentage and absolute count of circulating memory B lymphocytes and plasma cells.
  11. [11]
    The Role of Adhesion Molecules in Multiple Myeloma - PubMed
    Cellular adhesion molecules are involved in the homing of malignant plasma cells to the bone marrow, the production of growth factors and the recirculation ...<|control11|><|separator|>
  12. [12]
    Plasma Cell Markers | Antibodies.com
    Aug 7, 2025 · Explore our guide on plasma cell markers, such as CD38, CD138, CD27 and CD79a, used to identify and characterize plasma cells in health and ...Missing: BCR | Show results with:BCR
  13. [13]
    B-cell activation by armed helper T cells - Immunobiology - NCBI - NIH
    T-cell dependent antibody responses require the activation of B cells by helper T cells that respond to the same antigen; this is called linked recognition.
  14. [14]
    Assessment of T-dependent and T-independent immune responses ...
    Oct 10, 2012 · During a T-D antigenic challenge, a small proportion of activated B cells differentiate into short-lived plasma cells within the T-cell regions ...
  15. [15]
    IL-21 and CD40L synergistically promote plasma cell differentiation ...
    After undergoing Ig somatic hypermutation and antigen selection, germinal center (GC) B cells terminally differentiate into either memory or plasma cells (PCs).
  16. [16]
    IL-21 drives expansion and plasma cell differentiation of ... - Nature
    May 1, 2018 · IL-21 can potently induce CD11chiT-bet+ B cells and promote the differentiation of these cells into Ig-secreting autoreactive plasma cells.
  17. [17]
    The Interplay of IL-21 and BAFF in the Formation and ... - Frontiers
    ... IL-4 in combination with anti-CD40L did lessen plasma cell differentiation. Inhibition of other individual cytokines was found to diminish IgM production ...
  18. [18]
    Transcription factor IRF4 controls plasma cell ... - PubMed - NIH
    Plasma cell differentiation required IRF4 as well as the transcriptional repressor Blimp-1, which both acted 'upstream' of the transcription factor XBP-1.
  19. [19]
    Graded Expression of Interferon Regulatory Factor-4 Coordinates ...
    Plasma cell differentiation is regulated by the zinc-finger transcription factor Blimp-1 (encoded by the Prdm1 gene) and involves a dramatic expansion of ...
  20. [20]
    B cell memory: building two walls of protection against pathogens
    Dec 13, 2019 · We describe the two-phase process by which antigen drives the generation of long-lived plasma cells and memory B cells and highlight the challenges for ...Phase 1 Of B Cell Memory · Impact Of Antigen Structure · Phase 2 Of B Cell Memory
  21. [21]
    Class Switch Recombination Occurs Infrequently in Germinal Centers
    Here we have used multiple approaches to show that CSR is triggered prior to differentiation into GC B cells or plasmablasts and is greatly diminished in GCs.
  22. [22]
    Origin and Function of Circulating Plasmablasts during Acute Viral ...
    Indeed, the 6- to 7-day lag phase between memory B cell activation and plasma blast formation might be important to generate a minimal number of infected cells ...
  23. [23]
    Plasma cells: you are what you eat - PMC - PubMed Central
    Plasma cells are terminally differentiated B lymphocytes that constitutively secrete antibodies. These antibodies can provide protection against pathogens, ...
  24. [24]
    Multiple Levels of Immunological Memory and Their Association with ...
    First, short-lived plasma cells are formed upon contact with antigens and secrete low-affinity immunoglobulin M (IgM) antibodies for a few days, followed by ...Missing: Fas | Show results with:Fas
  25. [25]
  26. [26]
    The Fas antigen is detected on immature B cells and the ... - PubMed
    Except for the Burkitt cell lines and one plasma cell line, susceptibility to Fas-mediated apoptosis depended on Fas expression. Bcl-protein was detected on all ...
  27. [27]
    Dynamic organization of the bone marrow plasma cell niche - Aaron
    Feb 3, 2022 · In this review, we focus on how known factors regulate the longevity of plasma cell fitness and survival, and how that landscape is shaped by environmental ...Abstract · Introduction: an ode to the... · Conclusion: how PC dynamics...
  28. [28]
    APRIL is critical for plasmablast survival in the bone marrow and ...
    Mar 1, 2008 · We demonstrate here that APRIL is a critical factor for the establishment of the adult BM reservoir of anti-TT IgG-secreting cells.
  29. [29]
    Bone Marrow Tests for Multiple Myeloma | The IMF
    Normal bone marrow has about 2% or fewer plasma cells. The presence of 60% or more plasma cells in the bone marrow is an independent myeloma-defining event.
  30. [30]
    Repression of the Transcription Factor Bach2 Contributes to ...
    Jul 25, 2013 · Our results suggest that repression of the Bach2 transcription factor, which results from antigen experience, contributes to predisposition of IgG1 memory B ...
  31. [31]
    Plasma Cell Survival Is Mediated by Synergistic Effects of Cytokines ...
    In this study, we show that the cytokines IL-5, IL-6, TNF-α, and stromal cell-derived factor-1α as well as signaling via CD44 support the survival of isolated ...
  32. [32]
    Generation of human long-lived plasma cells by developmentally ...
    Dec 24, 2021 · Furthermore, Hammarlund et al (2017) recently demonstrated the persistence of LLPC in the BM for 10 yr after tetanus vaccination, thereby ...<|control11|><|separator|>
  33. [33]
    Hypoxia and hypoxia-inducible factor signals regulate the ... - Frontiers
    This review focuses on recent discoveries about how hypoxia and HIF signaling regulate the development, metabolism, and function of B cells.
  34. [34]
    Biochemical coordination of plasma cell genesis - PMC
    Indeed, past studies suggest that the average plasma cell secretes some 10,000 antibody molecules every second. The secretion of copious amounts of antibodies ...
  35. [35]
    Plasma cell immunoglobulin M molecules. Their biosynthesis ...
    (2) H and L chain assembly occurs between nascent H chains and a pool of free light chains present in the RER, followed by interchain disulfide bonding and ...
  36. [36]
    The role of N-glycosylation in B-cell biology and IgG activity. The ...
    Plasmocytes, as the final stage of B cell differentiation, produce and secrete immunoglobulins (Igs), of which IgGs are the most abundant N-glycosylated ...
  37. [37]
    Immunoglobulin class‐switch recombination: Mechanism, regulation ...
    According to the different regions of the Ig H chain, Ig is classified into five isotypes, namely IgM, IgG, IgA, IgE, and IgD. IgM typically exists in ...
  38. [38]
    XBP1, downstream of Blimp-1, expands the secretory apparatus and ...
    The differentiation of B cells into immunoglobulin-secreting plasma cells is controlled by two transcription factors, Blimp-1 and XBP1.
  39. [39]
    Plasma cell differentiation initiates a limited ER stress response by ...
    Plasma cell differentiation initiates a limited ER stress response by specifically suppressing the PERK-dependent branch of the unfolded protein response.
  40. [40]
    quantifying antibody secretion from individual plasma cells - PubMed
    Jul 31, 2009 · Using this technique we were able to determine that plasma cells recovered from the peripheral blood secreted the most antibody (1.667 ng/cell/ ...
  41. [41]
    The Humoral Immune Response - Immunobiology - NCBI Bookshelf
    The humoral immune response is mediated by antibody molecules that are secreted by plasma cells. Antigen that binds to the B-cell antigen receptor signals B ...
  42. [42]
    Physiology, Immune Response - StatPearls - NCBI Bookshelf - NIH
    Antibody production: Activated B cells differentiate into plasma cells, producing antibodies that neutralize pathogens or mark them for destruction. The ...
  43. [43]
    Physiology, Opsonization - StatPearls - NCBI Bookshelf
    Opsonins are used to overcome the repellent force between the negative cell walls and promote uptake of the pathogen by the macrophage. Opsonization is an ...
  44. [44]
    Persistence of long-lived plasma cells and humoral immunity ... - NIH
    These results provide strong evidence for the existence of memory B-cell–independent, long-lived PCs in humans that contribute to long-lasting humoral immunity.
  45. [45]
    Survival of long-lived plasma cells is independent of antigen - PubMed
    Our results confirm that persistent antibody titers are provided by long-lived PC, independent of memory B cells and demonstrate that this humoral memory is ...
  46. [46]
    The distribution and functions of immunoglobulin isotypes - NCBI - NIH
    It is believed that the primary functional role of IgA antibodies is to protect epithelial surfaces from infectious agents, just as IgG antibodies protect the ...
  47. [47]
    Biochemistry, Immunoglobulin A - StatPearls - NCBI Bookshelf - NIH
    May 1, 2023 · Immunoglobulin A (IgA), one of the five primary immunoglobulins, plays a pivotal role in mucosal homeostasis in the gastrointestinal, respiratory, and ...
  48. [48]
    Cellular and Chromatin Dynamics of Antibody-Secreting Plasma Cells
    The plasma cell differentiation program can be triggered by different signals and in multiple, diverse peripheral B cell subsets. This heterogeneity raises ...
  49. [49]
    The origins of vertebrate adaptive immunity - PMC - PubMed Central
    In the course of a humoral immune response, B cells undergo terminal differentiation into plasma cells, which secrete large amounts of soluble immunoglobulins.
  50. [50]
    Survival of Long-Lived Plasma Cells (LLPC): Piecing Together the ...
    Longevity is the hallmark of LLPC, but why and how they survive and function for years after antigen exposure is only beginning to be understood.Llpc And Memory B Cells Are... · Intrinsic Signals For Llpc... · T Regulatory Cells
  51. [51]
    Long-term immunogenicity after measles vaccine vs. wild infection
    The development of long-lived plasma cells is necessary to sustain life-long plasma antibody levels. In Italy, a single-antigen measles vaccine was introduced ...
  52. [52]
    Multiple Myeloma - StatPearls - NCBI Bookshelf
    Multiple myeloma (MM) is a clonal plasma cell proliferative disorder characterized by the abnormal increase of monoclonal immunoglobulins.
  53. [53]
    Impact of CRAB Symptoms in Survival of Patients with Symptomatic ...
    The acronym CRAB summarizes the most typical clinical manifestations of multiple myeloma, these being hypercalcemia, renal failure, anemia, and bone disease.Missing: plasma | Show results with:plasma
  54. [54]
    Current risk stratification and staging of multiple myeloma ... - Nature
    Jul 23, 2025 · MGUS is defined by the presence of a circulating monoclonal (M) protein <3 g/dL, clonal bone marrow plasma cells (BMPCs) < 10%, and no evidence ...
  55. [55]
    Renal Manifestations of Plasma Cell Disorders
    Waldenström macroglobulinemia is associated with an immunoglobulin M (IgM) monoclonal protein, and can cause hyperviscosity syndrome, anemia, lymphadenopathy, ...
  56. [56]
    AL amyloidosis, Waldenström macroglobulinemia, and POEMS ...
    AL amyloidosis typically develops from the background of a plasma cell neoplasm but can be associated with other lymphoproliferative disorders in which there ...
  57. [57]
    Plasma Cell Differentiation Pathways in Systemic Lupus ...
    Mar 5, 2018 · Plasma cells (PCs) are responsible for the production of protective antibodies against infectious agents but they also produce pathogenic antibodies in ...
  58. [58]
    Autoantibodies in Rheumatoid Arthritis: Historical Background ... - NIH
    Autoantibodies represent a hallmark of rheumatoid arthritis (RA), with the rheumatoid factor (RF) and antibodies against citrullinated proteins (ACPA) being ...
  59. [59]
    Common Variable Immunodeficiency - StatPearls - NCBI Bookshelf
    Common variable immunodeficiency is a disorder characterized by defective B cell function leading to impaired immunoglobulin production.
  60. [60]
    Gastrointestinal Disorders Associated with Common Variable ...
    Tissue histology in CVID is characteristic for the paucity or absence of plasma cells and a high frequency of CD8+ T cell infiltrates in the intestinal lamina ...
  61. [61]
    Agammaglobulinemia: X-linked (XLA) and autosomal recessive (ARA)
    People with genetic causes of low or absent antibodies (agammaglobulinemia) have a severe form of antibody deficiency with absent B cells.
  62. [62]
    Understanding and Interpreting Serum Protein Electrophoresis - AAFP
    Jan 1, 2005 · Diagnosis requires 10 to 15 percent plasma cell involvement on bone marrow biopsy. Anemia, pancytopenia, hypercalcemia, and renal disease ...
  63. [63]
    Tests for Multiple Myeloma | American Cancer Society
    Feb 28, 2025 · A diagnosis of multiple myeloma requires: Either a plasma cell tumor (proven by biopsy) OR at least 10% plasma cells in the bone marrow, AND at ...
  64. [64]
    Plasma Cell Neoplasms (Including Multiple Myeloma) Treatment ...
    Apr 25, 2025 · A bone marrow examination from an uninvolved site contains less than 10% plasma cells.[39-41] The absence of plasma cells on flow cytometry of ...
  65. [65]
  66. [66]
    Autoantibodies in COVID-19 survivors with post-COVID symptoms
    Jul 5, 2024 · Although evidence suggests that persistent autoantibodies can be associated with post-COVID symptoms, the clinical relevance of their presence seems modest at ...
  67. [67]
    Prevalent and persistent new-onset autoantibodies in mild to severe ...
    Oct 17, 2024 · We found an increased prevalence of new-onset autoantibodies after severe COVID-19 and demonstrated associations between distinct new-onset ...
  68. [68]
    Why Proteasome Inhibitors Cannot ERADicate Multiple Myeloma - NIH
    Plasmablasts and plasma cells are sensitive to proteasome inhibitors such as bortezomib because of their high level of immunoglobulin synthesis, leading to ...
  69. [69]
    Response of myeloma to the proteasome inhibitor bortezomib is ...
    Jan 1, 2012 · The view that myeloma is inherently sensitive to proteasome inhibitors because of its dependence on the UPR is satisfying, being consistent ...
  70. [70]
    Predicting the response of multiple myeloma to the proteasome ...
    Jun 10, 2016 · Proteasome inhibitors (PI) such as Bortezomib have increased the response rate and survival of patients with MM. The overall patient response ...Missing: review | Show results with:review
  71. [71]
    Targeting CD38 with Daratumumab Monotherapy in Multiple Myeloma
    Aug 26, 2015 · Daratumumab monotherapy had a favorable safety profile and encouraging efficacy in patients with heavily pretreated and refractory myeloma.
  72. [72]
    Daratumumab: a first-in-class CD38 monoclonal antibody for the ...
    Jun 30, 2016 · Daratumumab is a human monoclonal antibody that targets CD38, a cell surface protein that is overexpressed on multiple myeloma (MM) cells.
  73. [73]
    Treatment of multiple myeloma with the immunostimulatory SLAMF7 ...
    The primary mechanism of action of elotuzumab against myeloma cells is NK cell-mediated ADCC (B). Elotuzumab also activates NK cells directly via SLAMF7 binding ...
  74. [74]
    Elotuzumab Therapy for Relapsed or Refractory Multiple Myeloma
    Jun 2, 2015 · SLAMF7 expression is highest on plasma cells (malignant and normal), natural killer cells, and a subgroup of other immune cells, with no ...
  75. [75]
    Lenalidomide enhances anti-myeloma cellular immunity - PMC
    Lenalidomide is an effective therapeutic agent for multiple myeloma that exhibits immunomodulatory properties including the activation of T and NK cells.Missing: clearance | Show results with:clearance
  76. [76]
    Lenalidomide use in multiple myeloma (Review)
    Nov 28, 2023 · Lenalidomide reduces the immune checkpoint inhibitor programmed death-1 (PD-1) expression on both T and NK cells in patients with MM; it also ...
  77. [77]
    Immunomodulatory drugs in multiple myeloma - PubMed Central - NIH
    Lagrue et al. reported that lenalidomide lowers the threshold for NK cell activation and increases the amount of IFNγ production in stimulated cells [139].
  78. [78]
    FDA approves idecabtagene vicleucel for multiple myeloma
    Mar 29, 2021 · On March 26, 2021, the Food and Drug Administration approved idecabtagene vicleucel (Abecma, Bristol Myers Squibb) for the treatment of adult ...
  79. [79]
    Idecabtagene Vicleucel in Relapsed and Refractory Multiple Myeloma
    Feb 24, 2021 · Nearly all tumors expressed BCMA, a finding that supports the use of ide-cel in relapsed and refractory myeloma without restriction based on ...<|separator|>
  80. [80]
    Bispecific Antibodies in Multiple Myeloma: Present and Future - PMC
    Bispecific antibodies are a novel immunotherapeutic approach designed to bind antigens on malignant plasma cells and cytotoxic immune effector cells.
  81. [81]
    Article CD138 and APRIL regulate plasma cell survival, competition ...
    Aug 26, 2025 · APRIL and BAFF co-blockade depletes BM PCs within days to weeks after blockade initiation.,, While BAFF inhibition ...
  82. [82]
    Long-Lived Plasma Cells in Autoimmunity: Lessons from B ... - NIH
    Dec 27, 2013 · Rituximab achieves almost complete peripheral B-cell depletion, with minimal numbers of CD19+ cells being detectable in blood within 6 months ...
  83. [83]
    Rituximab specifically depletes short-lived autoreactive plasma cells ...
    B cell-depletion therapy through rituximab has already been shown to be effective in rheumatoid arthritis, multiple sclerosis, and several other autoimmune ...
  84. [84]
    The role of plasma exchange in the management of autoimmune ...
    The main mechanisms of action of TPE include the removal of circulating autoantibodies, immune complexes, complement components, cytokines and adhesion ...
  85. [85]
    Plasma Exchange (Plasmapheresis)
    Plasma exchange helps by removing these abnormal antibodies from the blood. Plasma exchange involves removing a person's blood drawn and putting their blood ...
  86. [86]
    mRNA vaccines induce durable immune memory to SARS-CoV-2 ...
    mRNA vaccines induce robust germinal center responses in humans (14, 15), which result in memory B cells that are specific for both the full-length SARS-CoV-2 ...
  87. [87]
    SARS-CoV-2 specific plasma cells acquire long-lived phenotypes in ...
    Basic mRNA vaccination with 3–4 doses are sufficient to induce memory plasma cells in man. Antigen-specific humoral immunity is based on an array of plasma ...