Fact-checked by Grok 2 weeks ago

B1 cell

B-1 cells are a distinct subpopulation of B lymphocytes characterized by innate-like properties, including early embryonic origins, self-renewal capacity, and the spontaneous production of polyreactive natural antibodies that provide constitutive protection against pathogens and aid in apoptotic cell clearance. Unlike conventional B-2 cells, which rely on T cell-dependent activation and undergo affinity maturation in germinal centers, B-1 cells exhibit blunted B cell receptor signaling, T cell-independent responses, and a less diverse repertoire biased toward germline-encoded antibodies. In mice, B-1 cells are phenotypically defined by high CD19 and IgM expression, low B220 and IgD levels, CD43 positivity, and the absence of CD23, with further subdivision into B-1a (CD5+) and B-1b (CD5-) subsets that predominate in peritoneal and pleural cavities. Their human counterparts, though less abundant and more heterogeneous, are typically identified by markers such as CD19+ CD20+ CD27+ CD43+ and low-to-intermediate CD38 expression, fulfilling functional criteria like natural antibody secretion and innate immune roles. B-1 cells develop in distinct waves during , primarily from fetal liver and extra-embryonic progenitors around embryonic day 9 in mice, bypassing the need for adult bone marrow-derived hematopoietic stem cells that generate B-2 cells. This early lineage commitment is regulated by transcription factors like Lin28b and Arid3a, enabling self-maintenance through proliferation in peripheral tissues such as the and , rather than recirculation or de novo generation. Transcriptional profiles further distinguish them, with constitutive ERK activation and impaired responses contributing to their innate bias. Functionally, B-1 cells are pivotal in basal , secreting low-affinity IgM natural antibodies that target conserved microbial motifs, oxidized lipids, and self-antigens to neutralize infections, promote of dead cells, and prevent . Beyond antibody production, they serve as efficient antigen-presenting cells, migrate to inflamed sites, and secrete regulatory cytokines such as IL-10 to dampen excessive and orchestrate . B-1b cells, in particular, provide adaptive-like recall responses to certain encapsulated , while the subset as a whole contributes to , protection against ischemia-reperfusion injury, and modulation of chronic conditions like and neurodegeneration. With aging, B-1 cell numbers and natural antibody output decline, correlating with increased susceptibility to infections and inflammatory diseases, underscoring their conserved role across vertebrates in bridging innate and adaptive immunity. Emerging research positions B-1 cells as potential therapeutic targets for enhancing vaccine responses or treating autoimmune disorders through their multifaceted regulatory functions.

Definition and Characteristics

Definition

B-1 cells represent a distinct subpopulation of B lymphocytes that primarily contribute to innate-like by producing polyreactive natural antibodies, such as IgM, in the absence of prior exposure. These antibodies provide constitutive protection against pathogens and maintain through broad reactivity to both self and foreign s. In contrast to conventional B-2 cells, which mediate adaptive immunity with antigen-specific responses and classical immunological , B-1 cells generally exhibit limited clonal expansion and, unlike conventional B-2 cells, do not form classical long-lived memory cells, though B-1b cells can provide some responses; they instead rely on rapid, T cell-independent activation to mount immediate defenses. This functional distinction positions B-1 cells as a bridge between innate and adaptive immunity, emphasizing their role in early-stage responses to infections. In adult mice, B-1 cells are predominantly enriched in coelomic cavities, including the peritoneal and pleural spaces, with additional populations residing in the and . They were first identified in the early as a unique subset of CD5+ (Ly-1+) B cells demonstrating self-renewal capabilities, distinct from the predominant B cell lineages. B-1 cells are further subdivided into B-1a and B-1b subtypes based on phenotypic and functional differences.

Key Features

[Condensed to avoid duplication: Focus on unique aspects like polyreactivity and residency, but since intro covers, minimize.] B-1 cells are distinguished by their long-lived nature and capacity for self-renewal in peripheral tissues, allowing them to maintain stable populations without reliance on continuous input. A hallmark of B-1 cells is their polyreactive repertoire, which recognizes conserved epitopes on microbial pathogens and self-antigens, facilitating broad, rapid protection. B-1 cells preferentially reside in tissue sites such as body cavities (peritoneal and pleural) and mucosal areas like the intestine, positioning them for frontline defense against pathogens entering through these barriers.

Origin and Development

Embryonic and Fetal Development

B1 cells are primarily generated during embryonic and fetal stages, with development peaking before birth in specific hematopoietic sites such as the fetal liver and omentum. In mice, B1 progenitors emerge as early as embryonic day 9.0–9.5 from the and para-aortic splanchnopleura, transitioning to the fetal liver where Lin⁻ CD93⁺ CD45R⁻/lo ⁺ cells predominate by day 17 of gestation. The fetal omentum serves as a selective niche for B1 cell production, supporting the biased generation of these cells over conventional lymphocytes. This fetal-restricted ontogeny contrasts with B2 cell development, which occurs predominantly in the adult . Early commitment to the lineage depends on distinct , including low expression of the identity factor Pax5 and high levels of Lin28b. Reduced Pax5 activity in fetal progenitors allows for an alternate developmental pathway that bypasses stringent B2-like checkpoints, facilitating the production of self-renewing B1 cells. Lin28b, highly expressed in fetal liver pro-B cells, promotes by repressing let-7 microRNAs and upregulating the Arid3a, which is sufficient to drive B1a cell generation even from adult pro-B precursors when ectopically expressed. This Lin28b-Arid3a axis ensures fetal-specific B cell fate decisions, with intact signaling required for full B1a differentiation. Postnatally, fetal-derived B1 progenitors migrate from these embryonic sites to peripheral tissues, such as the peritoneal and pleural cavities, to establish the lifelong cell pool. This migration occurs rapidly after birth, with transitional B1 cells entering the via the omentum. The rarity of new B1 production in adults stems from the insensitivity of B1 progenitors to bone marrow stromal signals, as demonstrated by the failure of adult to reconstitute B1 cells in transfer experiments, unlike neonatal liver cells which efficiently do so.

Adult Maintenance and Self-Renewal

In adult mice, the B1 cell population is primarily sustained through peripheral self-renewal rather than ongoing production from hematopoietic stem cells. This process occurs predominantly in tissue sites such as the peritoneal and pleural cavities, where B1 cells undergo homeostatic to maintain their numbers. Adoptive transfer experiments demonstrate that mature peritoneal B1 cells can reconstitute the entire B1 compartment in recipient mice, highlighting their capacity for self-renewal independent of central lymphoid organs. Recent studies have shown that transcription factors TCF1 and LEF1 are essential for B-1a cell , preventing excessive and exhaustion while maintaining IL-10 and expression. Key signals driving this homeostatic include B cell-activating factor (BAFF), which promotes B1 cell survival and expansion in the by differentially regulating inhibitory receptors like FcγRIIb. Additionally, IL-5 receptor signaling supports and self-renewal, particularly in response to local environmental cues in lipid-rich tissues, where enables metabolic adaptation for long-term persistence. The initial seeding of the B1 pool occurs during fetal and early postnatal stages, after which adult maintenance relies on these peripheral mechanisms. B1 cell progenitors exhibit stem-like properties that facilitate long-term repopulation, as evidenced by transplantation models where fetal-derived progenitors, supported by factors like Bmi1, maintain self-renewal and reconstitute the B1 compartment over extended periods. Bmi1 expression is essential for preserving this proliferative potential, with its deficiency leading to impaired maintenance of peritoneal B1 cells in competitive repopulation assays. These stem-like features underscore the innate-like, long-lived nature of B1 cells in adult . With advancing age, the efficiency of B1 cell renewal declines, contributing to through reduced repertoire diversity and functional potency. Although absolute B1 cell numbers remain relatively stable, the population shifts toward clonal dominance with increased mutations in receptors, diminishing the effectiveness of natural antibodies and heightening vulnerability to and chronic inflammation, as detailed in 2024 analyses of aging impacts. Environmental factors, including microbial signals from the , influence the maintenance of the B1 cell pool by modulating class-switch recombination and enhancing immunoglobulin diversity via activation and BAFF/ pathways. In germ-free conditions, B1 cells exhibit altered functional profiles, such as reduced IgA and IgG production, indicating that commensal microbes contribute to sustaining the qualitative integrity of the adult B1 compartment.

Classification and Subtypes

B1a Subtype

B1a cells, a major subset of B1 B cells in mice, are characterized by the surface markers CD5+, CD43+, and Mac-1+ (also known as CD11b+), along with high IgM expression, low IgD, low B220, and negative CD23. These cells represent a self-renewing that primarily originates during fetal and neonatal development and persists into adulthood through homeostatic proliferation. As major producers of germline-encoded natural IgM antibodies, B1a cells generate polyreactive immunoglobulins that target conserved self-antigens, such as phosphorylcholine on apoptotic cells and oxidized lipids. Their activation occurs through T-cell-independent pathways, enabling rapid IgM secretion in response to innate stimuli like pathogen-associated molecular patterns, which supports early defense and tissue homeostasis. In this capacity, B1a-derived natural IgM facilitates opsonization of dying cells, promoting their clearance by phagocytes and preventing the release of pro-inflammatory contents. Additionally, B1a cells exhibit regulatory functions by secreting IL-10, which suppresses excessive inflammation and maintains immune tolerance, particularly in contexts like autoimmune models and infection resolution. They are enriched in the spleen, peripheral blood, and coelomic cavities, where they constitute a significant proportion of circulating B cells. In humans, the putative equivalents of murine B1a cells are identified as CD20+CD27+CD43+ B cells, which display similar polyreactivity and produce natural antibodies with germline-like repertoires. However, the identification of distinct B1 cell subsets in humans remains controversial and is primarily based on phenotypic and functional similarities to murine cells. These cells are found in umbilical cord blood and adult peripheral blood, contributing to innate-like immunity through spontaneous IgM secretion against self and microbial antigens. Unlike B1b cells, which generate more diverse antigen-specific responses, B1a cells prioritize broad, low-affinity recognition via their restricted BCR repertoire.

B1b Subtype

B1b cells represent a distinct subset of B1 cells in mice, characterized by the surface phenotype CD5⁻ CD43⁺ B220ᵇᵒʷ IgMʰⁱ CD23⁻ IgDˡᵒʷ. Unlike B1a cells, which primarily produce natural antibodies against self-antigens, B1b cells exhibit a broader reactivity profile, responding to diverse microbial patterns such as bacterial and porins that extend beyond self-recognition. These cells predominate in the , where they constitute a significant portion of the B cell population and contribute to innate-like immune surveillance in body cavities. A key feature of B1b cells is their capacity for , enabling the generation of higher-affinity antibodies in response to T cell-independent type 2 antigens, including from pathogens. This process occurs in peritoneal B1b cells, particularly in association with IgA production, distinguishing them from other B1 subsets that show minimal in IgM-associated variable regions. Such adaptability allows B1b cells to mount rapid, extrafollicular responses without T cell help, enhancing against recurrent infections. B1b cells play a critical role in host defense against encapsulated bacteria, such as Streptococcus pneumoniae and Borrelia hermsii, by producing protective IgM antibodies that target conserved microbial structures like capsular and factor H-binding proteins. These responses include memory-like IgM production, providing long-term protection through sustained antibody levels and rapid recall upon re-exposure, as demonstrated in models of bacterial challenge and . This subset's specialization in T-independent immunity complements B1a functions, ensuring comprehensive coverage against polysaccharide-expressing pathogens. In humans, the B1b counterpart is proposed to be the CD20⁺ CD27⁺ CD43⁺ CD5⁻ CD70⁻ B cell subset, which shares innate-like properties and produces natural antibodies specific to polysaccharides of Streptococcus pneumoniae. However, the identification of distinct B1 cell subsets in humans remains controversial and is primarily based on phenotypic and functional similarities to murine cells. Their frequency declines with age, potentially impairing responses to encapsulated bacteria in older individuals.

Identification and Markers

Surface Markers in Mice

In mice, B1 cells are distinguished from conventional B2 cells by a characteristic surface phenotype that facilitates their identification in tissues such as the and . The core markers include high expression of and surface IgM (IgMhigh), coupled with low or absent expression of B220 (CD45Rlow/-) and positive expression of CD43. This phenotype reflects their innate-like properties and is consistently observed across studies using . B1 cells are subdivided into B1a and B1b subtypes based on differential expression of CD5 and CD11b, both of which express Mac-1 (CD11b). B1a cells exhibit high CD5 expression (CD5high CD11b+), while B1b cells are CD5-negative but CD11b-positive (CD5- CD11b+). CD43 positivity is shared by both subtypes, reinforcing their separation from B2 cells. To further discriminate B1 cells, markers typical of follicular B2 cells—such as high CD21 (CD21high) and (CD23+)—are excluded, as B1 cells typically lack these. The following table summarizes the key surface markers for B1 cells and their subtypes in mice:
MarkerB1 Cells (General)B1a SubtypeB1b SubtypeNotes/Exclusion from B2 Cells
High (+)High (+)High (+)Pan-B cell marker; essential for gating.
B220 (CD45R)Low/-Low/-Low/-B2 cells are B220high.
IgMHigh (+)High (+)High (+)Reflects constitutive antibody secretion.
(+)(+)(+)Shared marker; aids in distinction from B2.
CD5VariableHigh (+)-Defines B1a; modulates BCR signaling.
CD11b(+)(+)(+)Macrophage-like; present in both subtypes.
CD21/CD23Low/-Low/-Low/-B2 follicular cells are CD21high CD23+; excluded for B1.
CD5 expression on B1a cells is functionally linked to tonic B cell receptor (BCR) signaling, which supports cell survival and self-renewal by modulating inhibitory pathways, such as prolonged BCR-SHP-1 interactions that prevent excessive activation while maintaining basal homeostasis. This contrasts with stronger antigen-driven signaling in B2 cells.00530-6) For identification via flow cytometry, standard panels begin with gating on viable singlets using forward/side scatter, followed by selection of CD19+ cells to enrich for B cells. Subsequent analysis gates on B220low/- IgMhigh populations to isolate B1 cells, with CD43+ confirmation and exclusion of CD23+ or CD21high events to rule out B2 contamination. Subtype discrimination then uses a CD5 vs. CD11b plot, where B1a occupies the CD5high CD11b+ quadrant and B1b the CD5- CD11b+ quadrant. These panels effectively separate B1 cells from other leukocytes, such as T cells (CD3+) or myeloid cells (high CD11b without B markers), enabling precise quantification in murine models.

Markers in Humans

The identification of B1 cells in humans has presented significant challenges due to phenotypic and functional differences from their murine counterparts, where B1 cells are typically defined by and/or expression. In humans, proposed surface markers for B1 cells include + + + + -, with a notable lack of expression distinguishing them from B1 cells. These markers were established through flow cytometry-based sorting and functional validation in and peripheral blood samples. Additionally, human B1 cells exhibit low expression, further differentiating the primary population from rare + subsets that show higher levels. Functional confirmation of human B1 cells often relies on polyreactivity assays, which demonstrate their ability to produce antibodies binding multiple antigens with low affinity, a hallmark of innate-like B cell responses. These assays, involving ELISA or HEp-2 cell binding tests, confirm the polyreactive nature of antibodies secreted by sorted CD20+ CD27+ CD43+ CD70- B cells, aligning with their role in natural antibody production. Human B1 cells are enriched in specific tissues, including the spleen and umbilical cord blood, where they constitute a higher proportion of B lymphocytes compared to peripheral blood in adults. In peripheral blood, B1 cell frequency declines progressively with age, dropping from approximately 5-10% in newborns to less than 2% in the elderly, potentially contributing to reduced innate immunity in older individuals. The existence of a true B1 cell equivalent has been debated for years, with some studies questioning whether the proposed represents a distinct or overlaps with memory or activated B cells. However, recent single-cell RNA sequencing (scRNA-seq) studies from 2023 have confirmed the presence of putative human B1 cells in embryonic, fetal, and adult tissues, identifying transcriptional signatures akin to murine B1 cells, including self-renewal and innate genes. These findings, integrated across prenatal hematopoietic organs, resolve prior controversies by revealing B1-like clusters distinct from conventional cells.

Physiological Functions

Production of Natural Antibodies

B1 cells constitutively produce natural antibodies, predominantly low-affinity, polyreactive (IgM) that recognize conserved molecular patterns on both microbial and self-antigens. These antibodies arise from the limited variable () gene usage in B1 cell receptors, with specific segments such as the S107/TEPC15 preferentially encoding reactivity to phosphorylcholine (PC), a common component of bacterial cell walls, while VH11 or VH12 paired with Vκ9 targets (PtC), a self-lipid exposed on apoptotic cells. This -encoded repertoire enables broad, T cell-independent recognition without , ensuring rapid baseline protection against common threats. The secretion of these natural IgM antibodies occurs primarily from long-lived plasma cells differentiated from B1 cells, which reside in the , , and serosal cavities such as the and pleura. In mice, B1 cells account for over 80% of total serum IgM, maintaining circulating levels essential for steady-state immunity. B1a cells are the dominant subtype responsible for this production, contributing the majority of polyreactive IgM in naive animals. These natural antibodies fulfill critical homeostatic functions by neutralizing commensal bacteria in the gut and other mucosal sites, thereby limiting and promoting to the . Additionally, IgM specific to oxidized (oxLDL) enhances clearance of damaged lipids and apoptotic debris, reducing formation and attenuating progression in experimental models. Basal production of natural IgM by B1 cells proceeds independently of foreign exposure and is sustained through innate signaling pathways, including (TLR) engagement, which maintains differentiation and secretion without inducing adaptive responses. This regulation ensures continuous low-level output, supporting frontline defense and tissue throughout life.

Antigen Presentation and Phagocytosis

B1 cells exhibit notable phagocytic capacity, enabling them to engulf apoptotic bodies and pathogens, which contributes to tissue homeostasis and innate immune defense. This function is mediated primarily through their (BCR), as well as Fcγ receptors and scavenger receptors such as , which facilitate recognition and internalization of opsonized particles and oxidized lipids on apoptotic cells. Unlike follicular cells, B1 cells demonstrate higher efficiency in BCR-dependent phagocytosis, allowing rapid clearance of debris in peritoneal and splenic environments. In addition to phagocytosis, B1 cells serve as antigen-presenting cells (APCs) by expressing molecules, which process and present engulfed s to + T cells, thereby enhancing T-B interactions and supporting adaptive immunity. This MHC II-mediated presentation is particularly effective in co-stimulatory contexts, where B1 cells upregulate molecules like and to activate T helper cells. Studies indicate that B1a and B1b subtypes both contribute to this process, with B1b cells showing constitutive high MHC II expression for efficient antigen display. B1 cells further modulate immune responses through production, including IL-10 and TGF-β, which dampen excessive and promote tissue repair following infections. These s are secreted post-phagocytosis, fostering regulatory environments that resolve and support epithelial regeneration in infected tissues.

Immune Responses

Responses to Infections

B1 cells contribute to early defense against bacterial infections through T-independent type 2 (TI-2) responses triggered by repetitive antigens on surfaces, rapidly producing IgM antibodies without T cell assistance. In particular, B1a and B1b cells generate protective IgM targeting bacterial capsules, such as those of , which promotes opsonization, complement activation, and to limit dissemination. During infection, peritoneal B1 cells expand locally in response to pathogen-associated molecular patterns like LPS, undergoing and into antibody-secreting cells. Activated B1 cells then migrate from the peritoneal and pleural cavities to secondary lymphoid organs such as the , where they amplify IgM production and interact with other immune cells to enhance systemic control of the infection. In experimental models of sepsis induced by cecal ligation and puncture, B1a cells provide protection by secreting IgM and IL-10, which reduce excessive , bacterial load, and injury. Similarly, in models using S. pneumoniae, pleural B1a cells migrate to the and secrete polyreactive emergency IgM, improving survival and pathogen clearance. Studies from 2021 have further demonstrated cells' capacity to ameliorate outcomes in influenza virus and bacterial respiratory infections, often through rapid IgM-mediated neutralization and modulation of innate responses, highlighting their potential in severe pulmonary challenges. Despite these contributions, B1 cells show limited efficacy against viruses that depend on T cell help for affinity-matured, class-switched antibodies, as their responses remain predominantly T-independent and IgM-focused.

Role in Vaccination and Adaptive Immunity

B1b cells significantly enhance the to polysaccharide vaccines, such as the pneumococcal vaccine Pneumovax23, by recognizing native capsular from and mounting rapid, T cell-independent production. These cells expand robustly upon , differentiating into plasmablasts that secrete high levels of IgM and undergo class switching to IgG subclasses like IgG3 and IgG2b, resulting in elevated titers and improved opsonization for bacterial clearance. This memory-like responsiveness of B1b cells is particularly vital in older adults, where it compensates for waning adaptive immunity, providing long-lived protection against pneumococcal infections. B1 cells bridge innate and adaptive immunity through regulatory functions, such as IL-10 production that modulates adaptive responses by suppressing excessive T cell and pro-inflammatory release, thus aiding and preventing during humoral expansion. Concurrently, early secretion of polyreactive IgM opsonizes antigens and facilitates their uptake by antigen-presenting cells.

Pathological Implications

Involvement in Autoimmunity

B1 cells exhibit a dual role in , contributing to disease through the production of polyreactive while also exerting regulatory effects via anti-inflammatory cytokines. Their natural antibodies, which are typically low-affinity and germline-encoded, can recognize self-antigens and escape central and mechanisms, leading to autoreactive responses. In (RA), B1 cells are a of IgM (RF), an that binds the portion of IgG and promotes immune complex formation, exacerbating synovial . This polyreactivity arises from the limited BCR diversity of B1 cells, allowing recognition of multiple antigens including self-components like IgG, which drives chronic when tolerance checkpoints fail. In systemic lupus erythematosus (SLE), B1 cells are expanded and produce autoantibodies such as anti-phosphatidylserine (anti-PS) IgG, which deposit in renal glomeruli and activate TLR signaling via Syk , contributing to lupus progression. These antibodies overlap with homeostatic natural IgM functions but shift toward pathogenicity in dysregulated states, with reduced for self-antigens facilitating survival and expansion of autoreactive clones. Recent studies highlight B1a cells as key producers of PS-specific IgG in SLE patients, particularly those with , where levels correlate with disease severity. Conversely, cells, especially the B1a subset, regulate through IL-10 production, suppressing pro-inflammatory T cell responses. In experimental autoimmune (EAE), a mouse model of (), peritoneal B1a cells secrete IL-10 upon TLR stimulation, reducing Th1/Th17 activity and ameliorating disease severity. A 2022 review underscores B1 cells as innate-like regulators in , where their IL-10 dampens , with reduced B1-derived IL-10 linked to disease progression in patients. This regulatory function positions B1 cells as protective in early autoimmune stages. Therapeutic targeting of B1 cells reveals context-dependent outcomes, emphasizing their dual nature. In SLE models, selective depletion of B1a cells attenuates anti-PS production and slows , suggesting pathogenic dominance in established disease. However, in EAE, early B cell depletion—including regulatory B1 subsets—worsens clinical scores by eliminating IL-10-mediated suppression, highlighting the need for timed interventions to preserve beneficial functions. These findings from murine models inform potential strategies like IL-10 augmentation or subset-specific modulation in human .

Roles in Cancer and Other Diseases

B1 cells exhibit dual roles in cancer, contributing to both tumor progression and anti-tumor immunity. In solid tumors such as , regulatory B1a cells promote by secreting interleukin-10 (IL-10), which inhibits anti-tumor T responses and facilitates tumor growth. Similarly, peritoneal B1 cells enhance metastasis in models by homing to the omentum via signaling, where they support pre-metastatic niche formation through immunosuppressive production. These pro-tumor effects highlight B1 cells' capacity to modulate the adversely, as observed in 2024 studies emphasizing their infiltration into tumor sites and promotion of immune evasion. Conversely, B1 cells demonstrate anti-tumor potential through phagocytosis and production of natural IgM antibodies that target tumor-associated antigens. In leukemia models, B1-derived natural IgM recognizes glycolipids on malignant cells, inducing and enhancing tumor clearance. Additionally, B1 cells' phagocytic activity, mediated by signaling, allows uptake of tumor debris, potentially presenting antigens to augment adaptive responses, though this is more pronounced in early-stage models. (TLR4)-TRIF signaling in B1 cells further drives protective tumor-reactive IgM secretion, underscoring their role in innate surveillance against malignancies. In aging, the B1 cell pool declines, particularly in humans, leading to reduced natural antibody production and heightened susceptibility to infections. A 2024 review notes that this age-related impairs innate-like immunity, exacerbating to bacterial and pathogens in the elderly. Environmental pollutants contribute to this decline via immunotoxicity; for instance, exposure to chemicals and particles disrupts B1 cell , promoting and autoantibody production that further compromises immune function, as detailed in a 2023 analysis. Beyond cancer and aging, B1 cells play pathological roles in infectious diseases. In , increased B1 cell infiltration into Type 1 reaction lesions correlates with disease exacerbation through amplification of responses, according to a 2025 study on B cell subsets in lesions. Therapeutically, modulating B1a cells shows promise in ; their natural IgM production neutralizes viral components and clears debris, suggesting potential for enhancing B1a function to mitigate severe and in infected patients.

Laboratory Isolation and Study

Isolation Methods

B1 cells are primarily isolated from peritoneal cavity through peritoneal lavage, a that exploits their natural enrichment in this compartment. The begins by euthanizing the and injecting 5 ml of ice-cold () supplemented with 3% fetal calf serum (FCS) into the using a 27-gauge needle, followed by gentle and of the fluid with a 25-gauge needle. This yields approximately 5–10 million viable cells per , with 50–60% comprising B cells, of which a substantial proportion are B1 cells due to their preferential localization. To further purify mononuclear cells and remove debris or dead cells, the lavage fluid is subjected to density gradient centrifugation, such as using Lymphoprep or Percoll at 400–800 × g for 20–30 minutes, allowing collection of the interface layer containing enriched B1 cells. Post-isolation purity is typically assessed by , achieving >90% for B1 cells identified via markers like +CD5+CD11b+. Isolation of B1 cells from mouse spleen involves mechanical disruption to generate a single-cell suspension, followed by red blood cell (RBC) lysis to eliminate erythrocytes. The spleen is excised, placed in a dish with RPMI 1640 medium, and gently mashed through a 70-μm cell strainer using a plunger, then the suspension is centrifuged at 400–600 × g for 5 minutes. The pellet is resuspended in 2–5 ml of ACK lysis buffer and incubated for 3–5 minutes on ice to lyse RBCs, after which cells are washed and counted. This process yields a splenocyte population where B1 cells constitute 1–5% of total B cells (or <2% of splenocytes), reflecting their minor presence compared to the peritoneal cavity. For higher purity, magnetic-activated cell sorting (MACS) can be applied using anti-CD5 or anti-CD43 antibodies to positively select B1 cells, with flow cytometry confirming >90% purity based on CD19+CD5+CD43+ staining. In humans, B1 cells are isolated from sources such as or samples using density gradient centrifugation to obtain mononuclear cells. is diluted 1:1 with and layered over lymphocyte separation medium (density ~1.077 g/ml), then centrifuged at 1,500 × g for 20 minutes without brake to collect the interface, followed by washing at 500 × g. Splenic is similarly processed after mechanical dissociation and to generate a single-cell suspension before separation. These methods yield mononuclear fractions where B1 cells represent <1% to >9% of total B cells, depending on the donor. Purity exceeding 90% is achieved through subsequent flow cytometric sorting using markers such as CD20+CD27+CD43+CD3−, analyzed on instruments like BD LSR-II with appropriate controls.

Experimental Models and Techniques

Knockout mouse models have been instrumental in dissecting the developmental pathways of B1 cells. In CD5-deficient (CD5-/-) mice, B1 cells exhibit enhanced responsiveness to (BCR) crosslinking, displaying reduced and increased compared to wild-type counterparts, highlighting CD5's role in modulating B1 cell tolerance and activation thresholds. Similarly, conditional models reveal that loss of Pax5 in peripheral B lymphocytes leads to a significant reduction in B1a cells, underscoring Pax5's necessity for maintaining B1 cell identity and preventing into other lineages. These genetic disruptions allow researchers to isolate the contributions of specific transcription factors and surface molecules to B1 cell without confounding effects from global developmental arrest. Adoptive transfer experiments further elucidate B1 cell self-renewal mechanisms. Transfer of purified peritoneal B1a cells into congenic recipient mice demonstrates their capacity to repopulate and sustain the B1a compartment long-term, confirming intrinsic self-renewal properties independent of input. In Bmi1-deficient models, adoptive transfers show impaired maintenance of fetal-derived B1a cells, indicating Bmi1's pivotal role in preserving their proliferative potential. These assays provide quantitative insights into homeostatic regulation, with transferred cells often comprising over 80% of the recipient's peritoneal B1 pool after several months. In vitro assays enable precise evaluation of B1 cell activation and secretory functions. BCR crosslinking using anti-IgM antibodies stimulates B1 cell and production, revealing regulatory pathways such as those involving Lyn , which dampens excessive signaling to prevent . The enzyme-linked immunospot () assay quantifies spontaneous IgM secretion by peritoneal B1 cells, typically detecting 600-1,400 spot-forming units per 10^4 cells, and has been used to demonstrate differential secretion rates between B1a and B1b subsets. These techniques, often combined with , allow functional assessment of isolated B1 cells under controlled conditions. Recent advances in single-cell RNA sequencing (scRNA-seq) have uncovered transcriptional heterogeneity within B1 cell populations. scRNA-seq analyses of peritoneal B1 cells from young and aged mice identify distinct developmental trajectories, with aged cells showing clonal expansion and upregulated genes associated with , such as . These 2022 studies highlight two primary B1 lineages—one fetal-derived and self-renewing, the other adult-generated with limited persistence—providing a molecular map of heterogeneity. More recent 2024–2025 scRNA-seq studies, including those revealing TCF1 and LEF1's roles in B1a cell and exhaustion, further delineate subset-specific trajectories and regulatory networks. Complementing this, -Cas9 editing facilitates functional validation by targeting genes like (Blimp-1) in primary B cells, confirming its essential role in IgM secretion without altering B1 cell survival. In B1-enriched cultures, CRISPR knockouts of metabolic regulators, such as those in pathways, abolish self-renewal, validating scRNA-seq-derived candidates. Emerging techniques combine screens with scRNA-seq to dissect B1 cell functions, such as 2025 studies using pooled CRISPRi to reveal metabolic regulators of . Additionally, maps B1 interactions in tissues, enhancing understanding of their roles. visualizes dynamic B1 cell behaviors in the peritoneum during infection. Time-lapse imaging of whole-mount peritoneal tissues reveals B1 cell migration toward infection sites, such as in models of bacterial , where they cluster around pathogens within hours, facilitating and antibody deposition. These studies, often using fluorescently labeled anti-CD19 and anti-IgM antibodies, demonstrate B1 cells' rapid morphological changes, including pseudopod extension, during interactions with omental macrophages, underscoring their role in early immune .

References

  1. [1]
    The immunology of B-1 cells: from development to aging - PMC
    Aug 2, 2024 · B-1 cell: a unique innate-like lymphocyte. The B cell compartment is recognized by its clonally diverse population, bearing receptors that ...
  2. [2]
    B-1 Cell Development and Function - PMC
    May 11, 2015 · Roles of innate-like B cells in humoral immunity ... B-1 B cells are chiefly responsible for maintaining preimmune humoral defense at homeostasis.
  3. [3]
    Human B-1 Cells and B-1 Cell Antibodies Change With Advancing ...
    Mar 19, 2019 · In humans, B-1 cells have been identified as CD20+CD27+CD38low/intCD43+ based on the fact that B cells with this phenotype fulfill key ...
  4. [4]
    An Overview of B-1 Cells as Antigen-Presenting Cells - PMC - NIH
    Apr 11, 2016 · B-1 cells have peculiar features, such as a mixture of innate and lymphoid cell properties. As mentioned here, some authors describe ...
  5. [5]
  6. [6]
    The immunology of B-1 cells: from development to aging
    Aug 2, 2024 · B-1 cells have intricate biology, with distinct function, phenotype and developmental origin from conventional B cells.
  7. [7]
  8. [8]
    A Hard(y) look at B-1 cell development and function* - PMC
    Nov 15, 2018 · B-1 cells were shown to regulate immunity by cytokine secretion. A subset of body cavity B-1 cells appears to constitutively express IL-10, ...
  9. [9]
    The role of B-1 cells in inflammation | Immunologic Research
    Oct 1, 2015 · B-1 cells spontaneously secrete germline-like, repertoire-skewed polyreactive natural antibody, which acts as a first line of defense by ...
  10. [10]
    Protective Roles of Natural IgM Antibodies - PMC - PubMed Central
    Natural IgM antibodies that recognize apoptotic cells have been shown to enhance the phagocytic clearance of dead and dying cells and to suppress innate immune ...
  11. [11]
    Oxidation-specific epitopes are dominant targets of innate ... - JCI
    Apr 13, 2009 · B-1 cell–derived natural IgM Abs recognize oxidation-specific epitopes on apoptotic cells and in atherosclerotic lesions. Oxidation-specific ...
  12. [12]
    Homeostatic role of B-1 cells in tissue immunity - Frontiers
    B-1 B cells are a unique subset of B cells with innate-like properties, enriched in murine pleural and peritoneal cavities and distinct from conventional B-2 ...
  13. [13]
  14. [14]
    Lin28b promotes fetal B lymphopoiesis through the transcription ...
    Specifically, the transcription factor Arid3a, induced by Lin28b and a target of Let-7 miRNA, is sufficient to recapitulate fetal B cell development from bone ...Missing: omentum Pax5
  15. [15]
    Homeostatic role of B-1 cells in tissue immunity - PMC
    Sep 8, 2023 · Here we discuss B-1 cells, a subset of B cells with “innate-like” properties that have several features ascribed to tissue-resident lymphocytes.Figure 1 · Figure 2 · Figure 3<|control11|><|separator|>
  16. [16]
    FcgammaRIIb and BAFF differentially regulate peritoneal B1 cell ...
    May 15, 2013 · To identify possible receptors that differentially control B1 cell survival, we studied the expression of FcgammaRIIb on different B cell ...
  17. [17]
    Bmi1 Maintains the Self-Renewal Property of Innate-like B ... - PubMed
    Jun 15, 2020 · Our work has indicated that Bmi1 plays pivotal roles in self-renewal and maintenance of fetal-derived B-1a cells.
  18. [18]
    [PDF] B cells in the aging immune system: time to consider B-1 cells
    Role of B-1–derived natural IgM. B-1a cells are phenotypically characterized by cell surface markers: CD5+ IgMhigh IgDlow. B220low MAC-1+ CD23− CD43+.60 B-1 ...<|separator|>
  19. [19]
    Human B-1 cells take the stage - PMC - NIH
    B-1 cells differ from resting B-2 cells in a number of characteristics beyond surface antigen expression, natural antibody secretion, chronic intracellular ...
  20. [20]
    B-1a cells acquire their unique characteristics by bypassing the pre ...
    Oct 18, 2019 · B-1a cells are long-lived, self-renewing innate-like B cells that predominantly inhabit the peritoneal and pleural cavities.Introduction · Results · Igll1 And Wild-Type B-1a...
  21. [21]
    Nur77 Is Upregulated in B-1a Cells by Chronic Self-Antigen ...
    Nov 1, 2017 · B-1a cells are a unique population of innate-like B cells with a highly restricted and self-reactive BCR repertoire. Preimmune “natural” IgM ...
  22. [22]
    The Role of B-1 Cells in Inflammation - PMC - PubMed Central
    Abstract. B-1 lymphocytes exhibit unique phenotypic, ontogenic, and functional characteristics that differ from the conventional B-2 cells.
  23. [23]
    B-1 Cell Heterogeneity and the Regulation of Natural and Antigen ...
    Adoptive transfer of adult-derived peritoneal cavity B-1 cells into newborn mice seeds all major B-1 cell niches in spleen, body cavities, and bone marrow, ...
  24. [24]
    Immune Tolerance to Apoptotic Self Is Mediated Primarily ... - Frontiers
    Jan 18, 2018 · Regulatory B1a cells specifically recognize and augment tolerance to apoptotic self via IL-10 and NAbs; but once activated, can also prevent autoimmune ...
  25. [25]
    Human B1 cells in umbilical cord and adult peripheral blood express ...
    Human B1 cells consist of CD20 + CD27 + CD43 + CD70 − cells bearing a skewed B cell receptor repertoire, and are present in umbilical cord and adult peripheral ...
  26. [26]
    Pivotal Advance: Peritoneal cavity B-1 B cells have phagocytic and ...
    Upon phagocytosis, B-1a and B-1b cells were able to mature their phagosomes into phagolysosomes and displayed the ability to kill internalized bacteria.
  27. [27]
  28. [28]
  29. [29]
    Efficient B Cell Responses to Borrelia hermsii Infection Depend on ...
    Specifically, we have found that B1b cells are the dominant B cell subset that generates anti-B. hermsii- and anti-FhbA IgM responses. Additionally, we have ...
  30. [30]
    B-1 lymphocytes in mice and non-human primates - PMC
    B-1a and B-1b cells are mouse B cell populations with unique developmental, phenotypic, and functional characteristics. B-1b cells have been studied far less ...
  31. [31]
    Ikaros Is a Negative Regulator of B1 Cell Development and Function
    B1 cells are abundant in the peritoneal and pleural cavities where they can be further divided into B1a (CD19+CD5+CD11b+) and B1b (CD19+CD5−CD11b+) cells.
  32. [32]
    A differentiation pathway for B1 cells in adult bone marrow - PNAS
    Apr 7, 2009 · 2-step transplantation experiments established a differentiation pathway between conventional lymphoid progenitors, B1P, and mature B1 lymphocytes.
  33. [33]
    Role of CD5 in growth regulation of B-1 cells - PubMed - NIH
    CD5 promotes a prolonged interaction between BCR and SHP-1, which may be inhibitory to BCR signaling. CD5 was shown to modulate the function of autoantibody ...Missing: B1 tonic
  34. [34]
    Identify B Cell Subsets in Flow Cytometry (CD19, B220, Etc)
    Learn about the best flow cytometer parameters for the identification and analysis of B cells including plasma, B-1, B regulatory and splenic cells.<|control11|><|separator|>
  35. [35]
    Immunophenotyping | mouse B cells from spleen - Miltenyi Biotec
    The protocol uses flow cytometry to analyze B cell subsets (B1 and B2) in mouse spleen, identifying innate-like and conventional B cells.
  36. [36]
    Antigen Receptor Specificity and Cell Location Influence the ...
    Feb 1, 2021 · B-1a cells provide immediate and essential protection from infection through production of natural immunoglobulin, which is germline-like due to ...Elispot Assay · Dmpc And Dopc Elisa Analysis · Results
  37. [37]
    Selection at Multiple Checkpoints Focuses VH12 B Cell ...
    Phosphatidyl choline (PtC)-specific B cells segregate to the B-1 subset, where they comprise up to 10% of the B-1 repertoire. About half express VH12 and V.
  38. [38]
    Defining Natural Antibodies - Frontiers
    Jul 25, 2017 · The traditional definition of natural antibodies (NAbs) states that these antibodies are present prior to the body encountering cognate antigen.Natural Antibody (nab)... · Nab Characteristics · Defining Nabs
  39. [39]
    B-1 cells in the bone marrow are a significant source of natural IgM
    The study identifies populations of non-terminally differentiated B-1 cells in spleen and bone marrow as the most significant producers of natural IgM.
  40. [40]
    B-1 cells in immunotoxicology: Mechanisms underlying their ...
    Apr 18, 2023 · B-1 cells are already present and are active before the development of adaptive immunity. They produce natural antibodies with low affinity and ...
  41. [41]
    Naturally secreted immunoglobulins limit B1 and MZ B-cell numbers ...
    Jul 11, 2013 · Key Points. The study of AID−/−µS−/− mice reveals a microbiota-independent negative feedback control of MZ and B1 cell numbers by naturally ...
  42. [42]
    B-1b Cells Secrete Atheroprotective IgM and Attenuate Atherosclerosis
    Jun 16, 2015 · B-1-cell–mediated atheroprotection has been strongly linked to the production of atheroprotective IgM. In particular, IgM that bind OSE on OxLDL ...
  43. [43]
    TLR induces reorganization of the IgM-BCR complex regulating ...
    Aug 21, 2019 · Surface CD5 expression by B-1 cells has been linked previously to their inability to proliferate in response to BCR-mediated signaling (Bikah et ...<|control11|><|separator|>
  44. [44]
    Novel functions of murine B1 cells: Active phagocytic and ...
    Apr 24, 2012 · Our results reveal for the first time that mouse B1 cells have active phagocytic capabilities and thereby act as a bridge linking innate and adaptive immunity.<|control11|><|separator|>
  45. [45]
    B-cell subsets have different capacities for phagocytosis and ...
    Oct 13, 2025 · B cells are one of the 3 classic professional antigen-presenting cells (APCs) as defined by their baseline expression of both MHC class I and II ...B-Cell Subsets Have... · Results · Fo B Cells Present...<|separator|>
  46. [46]
    CD36 and LC3B initiated autophagy in B cells regulates the humoral ...
    Here we report that CD36 is involved in macro-autophagy/autophagy in B cells, and in its absence, the humoral immune response is impaired.Cd36 Controls... · Cd36 Interacts With Lc3b... · Cd36-Deficient B Cells Are...
  47. [47]
    The Other Function: Class II-Restricted Antigen Presentation by B ...
    In addition to their role as secretors of antibodies, B cells function as professional antigen-presenting cells (APCs) for CD4+ T cells by expressing cell- ...
  48. [48]
    Characterization of B-1b cells as antigen presenting cells in the ...
    Results showed that B1-b cells express constitutively high levels of class II MHC and costimulatory molecules inducing an efficient proliferation of gp43 ...
  49. [49]
    B cells support the repair of injured tissues by adopting MyD88 ...
    The B cell response to injury was multi-modal with simultaneous production of both regulatory cytokines such as IL-10, IL-35 and TGFβ, and inflammatory ...
  50. [50]
    The role of B-1 cells in cancer progression and anti-tumor immunity
    B-1 cells regulate anti-tumor immune responses and modulate tumor cell properties through contact-dependent and -independent mechanisms. (A) The interaction of ...
  51. [51]
    The role of B-1 cells in cancer progression and anti-tumor immunity
    Apr 2, 2024 · B-1 cells regulate anti-tumor immune responses and modulate tumor cell properties through contact-dependent and -independent mechanisms. (A) The ...
  52. [52]
    B-1a B Cells that Link the Innate and Adaptive Immune Responses ...
    B-1a cells are responsible for the production of IgM against thymus-independent antigens such as polysaccharides and phospholipids of the bacterial capsule (20 ...
  53. [53]
    B-1a and B-1b Cells Exhibit Distinct Developmental Requirements ...
    B-1a and B-1b lymphocytes were found to exhibit specialized roles in providing immunity to Streptococcus pneumoniae and differ dramatically in their ...
  54. [54]
    Regulation of B1 cell migration by signals through Toll-like receptors
    B1 cells are located mainly in the peritoneal and pleural cavities and express high levels of surface IgM and low levels of IgD, CD23, and B220. In addition to ...
  55. [55]
    GPR183 Is Dispensable for B1 Cell Accumulation and Function, but ...
    Previous studies have shown that in response to i.p. LPS injection, B1 cells migrate out of the peritoneal cavity through the omentum to the spleen and the gut, ...
  56. [56]
  57. [57]
    B-1a cells protect mice from sepsis-induced acute lung injury - NIH
    May 29, 2018 · Mice treated with B-1a cells showed dramatic improvement in lung injury compared to PBS-treated mice after sepsis. We found apoptosis in the ...
  58. [58]
    B-1a Cells Protect Mice from Sepsis: Critical Role of cAMP-response ...
    Jul 15, 2018 · Here we report that B-1a cells play a beneficial role in sepsis by mitigating exaggerated inflammation through a novel mechanism.
  59. [59]
    [PDF] Pleural innate response activator B cells protect against pneumonia ...
    Jul 12, 2013 · We show that in response to lung infection, B1a B cells migrate from the pleural space to the lung parenchyma to secrete polyreactive emergency.<|control11|><|separator|>
  60. [60]
    THERAPEUTIC POTENTIAL OF B-1A CELLS IN COVID-19 - NIH
    B-1a cells have been shown to ameliorate influenza virus infection, sepsis, and pneumonia, all of which are similar to COVID-19.
  61. [61]
    Aging Promotes B-1b Cell Responses to Native, but Not Protein ...
    Antibodies (Abs) against capsular polysaccharides (PPS) play a major role in promoting opsonization and clearance, and hence, current pneumococcal vaccines ...
  62. [62]
    B1b Cells Recognize Protective Antigens after Natural ... - Frontiers
    An unexpected benefit to studying B1b cells is that they appear to have a propensity to recognize protective antigens in bacteria.
  63. [63]
    IL-10+ innate-like B cells are part of the skin immune system and ...
    Innate-like B cells, in particular B1 cells, bridge innate and adaptive immunity by efficiently mounting rapid T cell-independent antibody (IgM and IgA) ...
  64. [64]
    B lymphocytes in COVID-19: a tale of harmony and discordance
    Apr 29, 2023 · Recent studies have revealed the critical protective and immune-regulatory role of natural IgM secretion by B-1 cells in response to viral ...
  65. [65]
    Hybrid immunity improves B cells and antibodies against SARS-CoV ...
    Oct 20, 2021 · Our data suggest that people who are seropositive following infection or primary vaccination will produce antibodies with increased potency and breadth.
  66. [66]
    Role of B-1a cells in autoimmunity - PubMed
    B-1a cells are largely responsible for the production of circulating IgM referred to as natural antibodies. These low affinity antibodies are polyreactive.
  67. [67]
    Implications for Induction of Autoimmunity via Activation of B-1 Cells ...
    Furthermore, the production of various B-1-cell-related autoreactive antibodies such as IgM-type rheumatoid factor, anti-single-stranded DNA antibody, and ...
  68. [68]
    B1-cell-produced anti-phosphatidylserine antibodies contribute to ...
    Jun 9, 2023 · Our study has demonstrated that the anti-PS autoantibodies produced by B1 cells contribute to lupus nephritis development.
  69. [69]
    Emerging role of innate B1 cells in the pathophysiology of ...
    B-1 cells are innate-like B cells that have features of both the adaptive and innate immune systems. •. The development of B-1 cells is distinct from that of ...
  70. [70]
    The Forgotten Brother: The Innate-like B1 Cell in Multiple Sclerosis
    Moreover, it was recently shown that the migration of B1 cells to the spleen after parasitic infection increases the percentage of T regulatory cells (Tregs). ...
  71. [71]
    A Regulatory B Cell Subset with a Unique CD1dhiCD5+ Phenotype ...
    May 16, 2008 · By contrast, B cell depletion early in the course of disease worsens EAE, whereas B cell depletion at the height of disease ameliorates EAE ...
  72. [72]
    Human B-1 cells are important contributors to the naturally-occurring ...
    ... natural tumor-reactive IgM production by B1 cells. Cancer Immunol Immunother ... leukemia cells. Glycoconjugate J (2013) 30(7):687–99. doi: 10.1007 ...
  73. [73]
    A TLR4-TRIF-dependent signaling pathway is required for protective ...
    A TLR4-TRIF-dependent signaling pathway is required for protective natural tumor-reactive IgM production by B1 cells. Cancer Immunol Immunother. 2020 Oct;69 ...
  74. [74]
    B-cell subsets in leprosy lesions: unraveling the complex interplay
    Aug 9, 2025 · To investigate the presence and distribution of B-cell subsets, including B1 cells, Marginal Zone (MZ) B-cells, Regulatory B-cells (Bregs), and ...
  75. [75]
    Isolation of Mouse Peritoneal Cavity Cells - PMC - NIH
    Jan 28, 2010 · Our protocol will describe a procedure for obtaining viable immune cells from the peritoneal cavity of mice, which then can be used for phenotypic analysis by ...Missing: density gradient
  76. [76]
    Mouse Spleen Cell Isolation Protocol | Thermo Fisher Scientific - US
    This mouse spleen cell isolation protocol outlines the steps for isolating splenocytes, which include various immune cell subtypes.
  77. [77]
    Homeostasis of Peripheral B Cells in the Absence of B Cell Influx ...
    Another B cell subset, consisting of the so-called B-1 cells, has self-renewing capacity and predominates in the peritoneal and pleural cavities 5. Recent data ...
  78. [78]
    B-1a Cell Isolation Kit, mouse | Miltenyi Biotec | USA
    The B-1a Cell Isolation Kit has been developed for the isolation of CD5+ B-1a cells from mouse body cavities or spleen. | Miltenyi Biotec | USA.
  79. [79]
    Isolation and Analysis of Human B1 Cells - PMC - NIH
    B1 cells constitute a distinct B cell lineage with a unique set of characteristics that includes constitutive production of natural antibody that is protective ...
  80. [80]
    CD5-Mediated Negative Regulation of Antigen Receptor ... - Science
    In CD5-deficient mice, B-1 cells responded to mIgM crosslinking by developing a resistance to apoptosis and entering the cell cycle.<|separator|>
  81. [81]
    Pax5 regulates B cell immunity by promoting PI3K signaling via ...
    The posttranscriptional downregulation of PTEN expression is an important function of Pax5 that facilitates the differentiation and survival of mature B cells.
  82. [82]
    B1a B cells require autophagy for metabolic homeostasis and self ...
    We show that B1 B cells are bioenergetically more active than B2 B cells, with higher rates of glycolysis and oxidative phosphorylation, and depend on ...Missing: seminal | Show results with:seminal
  83. [83]
    Bmi1 maintains the self-renewal property of innate-like B lymphocytes
    Finally, adoptive transfer of mature B-1a cells into congenic recipient mice was demonstrated to be sufficient to repopulate and maintain the B-1a cell ...
  84. [84]
    Multiple Regulatory Mechanisms Control B-1 B Cell Activation - PMC
    Dec 17, 2012 · In this review we summarize the mechanisms involved in regulation of BCR and Toll-like receptor (TLR) mediated B-1 cell activation.
  85. [85]
    Immunoglobulin secretion by B1 cells: Differential intensity and IRF4 ...
    Peritoneal B1 cells are typified by spontaneous, constitutive secretion of IgM natural antibody, detected by ELISPOT assay, among other means.
  86. [86]
    Single-cell genomics identifies distinct B1 cell developmental ...
    May 7, 2022 · B1 cells primarily reside in the peritoneal cavity and are known to originate from various fetal tissues, yet their developmental pathways and ...
  87. [87]
    In vivo analysis of CRISPR-edited germinal center murine B cells
    Oct 16, 2024 · We present a cost-effective, rapid, versatile, and adaptable CRISPR-Cas9 method for in vivo loss-of-function studies of individual murine B cell genes within ...
  88. [88]
    B‐1 cells are pivotal for in vivo inflammatory giant cell formation - 2005
    Jul 26, 2005 · As analysed by immunostaining and confocal microscopy, B-1 cells loose IgM expression in the course of differentiation keeping the expression of ...
  89. [89]
    Resident macrophage-dependent immune cell scaffolds drive anti ...
    Nov 9, 2021 · Here, we examined the mechanisms that control bacterial infection in the peritoneum using a mouse model of abdominal sepsis following ...