Fact-checked by Grok 2 weeks ago

B-cell activating factor

B-cell activating factor (BAFF), also known as B-lymphocyte stimulator (BLyS) or tumor necrosis factor ligand superfamily member 13B (TNFSF13B), is a belonging to the (TNF) superfamily that serves as a key regulator of B-cell survival, maturation, proliferation, and differentiation within the . Encoded by the TNFSF13B gene on human , BAFF is initially synthesized as a type II that undergoes proteolytic by to yield a soluble form, which predominantly assembles into homotrimers or, under physiological conditions, higher-order 60-mer structures to facilitate receptor binding. Discovered independently in 1999 by research groups led by Schneider et al. and Mackay et al., BAFF was quickly recognized for its essential role in maintaining B-cell , as its deficiency in mice leads to severe reductions in mature B cells and impaired . BAFF exerts its effects by binding to three distinct receptors on B cells and other immune cells: (BCMA), transmembrane activator and CAML interactor (TACI), and BAFF receptor (BAFF-R), with BAFF-R being the primary mediator of B-cell survival signals through pathways involving activation. Expressed mainly by innate immune cells such as monocytes, macrophages, dendritic cells, and neutrophils, as well as stromal cells in lymphoid tissues, BAFF supports the transition of immature B cells from the to the periphery and promotes the longevity of memory B cells and plasma cells, thereby enabling robust antibody production during immune responses. Beyond its canonical functions, emerging evidence highlights BAFF's broader influences, including contributions to T-cell regulation, innate immunity against infections, and pathological processes in and cancer. Dysregulated BAFF expression is implicated in numerous diseases, particularly those involving aberrant B-cell activity; elevated serum levels are observed in autoimmune disorders such as systemic lupus erythematosus (SLE), , and Sjögren's syndrome, where it sustains autoreactive B cells and exacerbates inflammation. In B-cell malignancies like (CLL) and (MM), BAFF fosters tumor cell survival and resistance to , correlating with disease progression and poor prognosis. Therapeutically, BAFF has become a prominent target, with —a that neutralizes soluble BAFF—approved by the FDA in 2011 for treating active SLE in adults and later for , demonstrating significant reductions in disease flares and steroid use in clinical trials. Ongoing research explores additional BAFF inhibitors, such as atacicept (a TACI-Ig ) and novel CAR-T therapies targeting BAFF receptors, in trials for autoimmune conditions and B-cell neoplasms, underscoring its potential as a trans-nosographic biomarker and intervention point in .

Discovery and nomenclature

Historical discovery

The B-cell activating factor (BAFF), also known as BLyS or TALL-1, was independently discovered in 1999 by four research groups through distinct approaches that highlighted its role in B-cell biology. Researchers at Sciences, led by et al., identified the protein as BLyS (B lymphocyte stimulator) via (EST) screening of a from activated monocytes, demonstrating its ability to induce B-cell proliferation and immunoglobulin secretion . Concurrently, et al. at Zymogenetics cloned TALL-1 (TNF- and ligand-related leukocyte-expressed ligand 1) using functional assays focused on factors enhancing B-cell survival, revealing its expression in T cells and downregulation by mitogens. Shortly thereafter, Schneider et al. at isolated BAFF based on to other members of the (TNF) family, confirming its potent stimulation of B-cell growth in culture. Additionally, Mukhopadhyay et al. identified THANK (TNF homologue that activates , NF-κB, and JNK) through screening for TNF family members with similar functional properties. These seminal studies, published in 1999, established BAFF as a novel TNF family critical for B-cell . Initial functional characterization rapidly followed the cloning efforts, with in vitro assays using recombinant BAFF protein and mouse splenic B cells showing enhanced in response to (LPS) or anti-IgM stimulation. Moore et al. reported that BLyS specifically promoted the survival and expansion of mature B cells while sparing other immune cell types, underscoring its selective action on the B-cell lineage. Similarly, Schneider et al. demonstrated dose-dependent B-cell growth promotion without inducing , linking BAFF to TNF family signaling motifs. These experiments, conducted in models, provided early evidence of BAFF's non-redundant role in supporting peripheral B-cell maintenance beyond central development stages. Early animal studies further validated BAFF's essential function, with the generation of BAFF-deficient mice in 2000-2001 revealing profound B-cell deficiencies. animals exhibited a near-complete absence of mature B cells in and lymph nodes, while early stages remained intact, indicating BAFF's necessity for transitional B-cell survival and maturation. This , coupled with reduced immunoglobulins, confirmed BAFF as a key survival factor, with implications for understanding B-cell-related immunodeficiencies.

Gene and nomenclature

The B-cell activating factor (BAFF) is encoded by the TNFSF13B gene, officially designated as TNF superfamily member 13b by the (HGNC). In humans, TNFSF13B is located on chromosome 13q33.3, spanning approximately 39 kb from positions 108,269,629 to 108,308,484 on the (GRCh38.p14). The mouse homolog, Tnfsf13b, resides on at position 8 A1.1 (4.55 cM). BAFF was independently identified in 1999 by multiple research groups, leading to initial names reflecting its discovery contexts: BAFF (B-cell activating factor) by Schneider et al., TALL-1 (TNF- and ligand-related leukocyte-expressed ligand 1) by et al., BLyS (B-lymphocyte stimulator) by et al., and THANK (TNF homologue that activates , , and JNK) by Mukhopadhyay et al.. These efforts culminated in the unified HGNC-approved symbol TNFSF13B, part of the , with an earlier provisional symbol TNFSF20; the preferred name BAFF and designation CD257 are widely used aliases, alongside BLyS (B-lymphocyte stimulator) and zTNF4. The TNFSF13B gene consists of 6 exons and is regulated by a promoter region containing at least one binding site, enabling transcriptional control by and NFAT factors.

Molecular structure

Gene organization

The TNFSF13B gene, located on 13q33.3, spans approximately 57 kb and consists of six s separated by five introns. Exon 1 encodes the cytoplasmic and transmembrane domains along with flanking regions, exon 2 includes the cleavage site essential for processing the membrane-bound form into a soluble , and exons 3 through 6 encode the extracellular domain featuring the TNF homology domain responsible for trimerization and receptor binding. The promoter region of TNFSF13B, spanning about 1 kb upstream of the transcription start site, contains regulatory elements including binding sites for transcription factors that modulate basal and inducible expression. Additionally, the promoter harbors interferon-responsive elements, such as an IRF-1 between -750 and -500 bp, which facilitate upregulation in response to type I interferons via JAK/STAT signaling. Intronic regions include potential enhancer elements that contribute to tissue-specific expression patterns, particularly in immune cells like monocytes and B lymphocytes. Evolutionary conservation of TNFSF13B is high across mammals, reflecting its critical role in B-cell ; for instance, the human protein shares 72% identity with the ortholog in the extracellular , with even greater conservation (over 85%) in the TNF core. No functional pseudogenes have been identified for TNFSF13B in humans or common model organisms. Several polymorphisms in TNFSF13B influence gene expression, notably the promoter variant rs9514828 (C>T at position -871), which is associated with increased transcript levels and elevated soluble BAFF protein in various immune contexts, though specific causal mechanisms remain under investigation.

Protein isoforms and assembly

B-cell activating factor (BAFF), also known as BLyS, is initially synthesized as a 285-amino-acid type II transmembrane glycoprotein precursor with an approximate molecular weight of 32 kDa. This precursor features an N-terminal cytoplasmic tail of 46 amino acids, a transmembrane domain spanning 21 amino acids, and a C-terminal extracellular domain comprising 218 amino acids. The extracellular region includes a TNF homology domain responsible for receptor interactions, flanked by a stalk region susceptible to proteolytic processing. The membrane-bound precursor is cleaved by a furin-like at the site RQKR ( 130–133) to generate the soluble isoform, which consists of 152 (from residue 134 to 285) and has a molecular weight of approximately 18 . This soluble form is predominantly secreted by monocytes and dendritic cells, allowing it to circulate and exert systemic effects on B cells. An alternative splice isoform, ΔBAFF, lacks 3 and results in a shorter protein that acts as a dominant-negative regulator by disrupting normal BAFF oligomerization and secretion. BAFF undergoes post-translational modifications, including N-linked glycosylation at Asn124 within the stalk region, as well as potential O-linked glycosylation at several serine/threonine residues in the extracellular domain. The N-glycosylation at Asn124 contributes to the molecular mass of the uncleaved precursor but is absent in the mature soluble form following proteolytic processing; this modification influences the stability of the membrane-bound form without altering the receptor-binding capability of the cleaved protein. In terms of assembly, the biologically active unit of BAFF is the homotrimer, formed through hydrophobic interactions within the TNF homology domain, with an approximate molecular weight of 60 kDa. Soluble BAFF trimers can further oligomerize into higher-order 60-mer structures (consisting of 20 trimers) under neutral or basic conditions, driven by trimer-trimer interactions mediated by the DE loop (also called the flap region) in the TNF domain, particularly involving at position 218. These 60-mer superclusters represent virus-like particles that enhance for BAFF receptors, though their formation is reversible and pH-sensitive, dissociating into trimers at acidic .

Biological functions

Role in B-cell maturation and survival

B-cell activating factor (BAFF), also known as B-lymphocyte stimulator (BLyS), plays an essential role in the survival of transitional B cells during their maturation from the to the . Immature B cells exiting the express BAFF receptor (BAFF-R) upon reaching the transitional stage, where BAFF binding to BAFF-R delivers critical anti-apoptotic signals that prevent . This interaction upregulates anti-apoptotic genes, particularly members of the such as and Mcl-1, thereby promoting the transition of these cells into mature naive B cells. In mature B cells, BAFF maintains by supporting the of follicular and marginal zone B cells in the . Through sustained BAFF-R signaling, BAFF ensures the survival and proper positioning of these subsets, which are vital for humoral immune responses. Additionally, BAFF contributes to by facilitating class-switch recombination (CSR) and immunoglobulin production, often in concert with other cytokines, thereby sustaining secretion over extended periods. The availability of BAFF establishes a for B-cell selection, where limited BAFF levels impose a competitive stringency that favors non-autoreactive clones while eliminating self-reactive ones during peripheral maturation. Excess BAFF disrupts this balance by rescuing autoreactive B cells from , potentially lowering the selection threshold and promoting their expansion. BAFF acts as a limiting resource, where its availability determines the size of the B-cell pool; insufficient levels lead to impaired B-cell pools. In BAFF mice, this deficiency manifests as profound reductions in mature B cells and , characterized by low serum IgM and IgG levels, underscoring BAFF's indispensable role.

Effects on other immune components

BAFF plays a significant role in modulating (DC) function beyond its primary effects on B cells. express BAFF and its receptors, and stimulation with BAFF promotes their maturation by upregulating co-stimulatory molecules such as and on myeloid DCs. This enhancement reduces phagocytic capacity while increasing production of inflammatory cytokines like IL-6 and chemokines such as and , thereby improving and facilitating Th1-biased immune responses. In addition to indirect influences through B-cell activation, BAFF directly modulates T-cell responses via low-affinity binding to receptors like BAFF-R expressed on T cells. This interaction promotes T-cell survival and proliferation by upregulating anti-apoptotic factors such as , and it can enhance differentiation toward Th1 phenotypes, particularly in inflammatory contexts where IFN-γ and TNF-α further induce BAFF expression. BAFF bridges innate and adaptive immunity by being produced by innate cells like neutrophils and macrophages during infections, where it amplifies humoral responses through B-cell survival and class-switch recombination signals. For instance, cytokine-activated neutrophils release BAFF in response to innate stimuli such as type I IFNs, supporting broader antibody production. BAFF also synergizes with , another TNF family member, to sustain survival via receptors like and TACI, with APRIL playing a more prominent role in early niches. Although primarily hematopoietic, BAFF exhibits limited expression in non-immune cells, including , where it acts as a proinflammatory contributing to . In obese models, BAFF promotes inflammation and , partly by facilitating B-cell infiltration into fat depots, which exacerbates metabolic dysfunction; depletion of BAFF improves glucose tolerance and reduces hepatic in aged mice.

Receptor interactions and signaling

Binding to receptors

B-cell activating factor (BAFF) interacts with three distinct receptors belonging to the receptor (TNFR) superfamily: BAFF-R (also known as TNFRSF13C or BR3), TACI (TNFRSF13B), and (TNFRSF17). These receptors are type I transmembrane proteins characterized by extracellular cysteine-rich domains (CRDs) that mediate binding. BAFF-R exhibits high specificity for BAFF, binding exclusively to it with a (Kd) of approximately 16 nM in monomeric form, though effects in bivalent contexts reduce this to less than 0.03 nM. In contrast, TACI and bind both BAFF and the related APRIL, with BAFF exhibiting a Kd of 0.1–0.3 nM to TACI and a weaker monomeric Kd of 1.6 μM to , which is enhanced to approximately 0.6 nM through multivalency. The distribution of these receptors across B-cell subsets reflects their specialized roles in B-cell homeostasis. BAFF-R is predominantly expressed on transitional type 2 and mature recirculating B cells in the periphery, but absent on plasma cells and centroblasts. TACI is found on marginal zone B cells, activated B cells, and plasma cells, with additional expression on activated T cells. BCMA is primarily restricted to long-lived plasma cells and a subset of memory B cells, where it supports survival in the and mucosal tissues. This differential expression ensures targeted BAFF signaling at key stages of B-cell development and maintenance. BAFF engagement occurs primarily through its homotrimeric form, which interacts with the CRD regions of the receptors to induce receptor clustering and signaling initiation. For BAFF-R, which contains a single CRD, to the soluble trimer is sufficient for activation. However, TACI (with two CRDs) and (with one CRD but lower intrinsic ) require higher-order assemblies, such as the 60-mer superclusters formed by membrane-bound or oligomeric BAFF, to achieve effective multivalency and stable . This multivalent presentation amplifies , particularly for TACI and , enabling robust responses in receptor-expressing cells. Unlike BAFF-R, the shared of to TACI and introduces ligand competition, but BAFF maintains exclusivity at BAFF-R due to structural differences in the ligand-receptor .

Downstream signaling pathways

Upon engagement of BAFF with its receptors BAFF-R, TACI, or BCMA, intracellular signaling is initiated primarily through recruitment of TNF receptor-associated factors (TRAFs), including TRAF2, TRAF3, TRAF5, and TRAF6, which serve as adaptor proteins to activate downstream cascades essential for B-cell survival, proliferation, and differentiation. The canonical NF-κB pathway is activated by all three receptors, though to varying degrees, involving the formation of a signaling complex with TRAF2, TRAF5, or TRAF6 that recruits the IκB kinase (IKK) complex, leading to phosphorylation and ubiquitin-mediated degradation of IκBα. This releases the p50/RelA (p65) heterodimer, which translocates to the nucleus to transcribe anti-apoptotic and survival genes such as Bcl-2 and A1. In BAFF-R and TACI, this pathway supports mature B-cell maintenance, while BCMA activation in plasma cells reinforces longevity. BAFF-R uniquely drives the non-canonical pathway through TRAF3-mediated polyubiquitination and degradation, which stabilizes -inducing () by preventing its turnover. Stabilized then phosphorylates IKKα, promoting processing of the p100 precursor to p52, which dimerizes with RelB and translocates to the to induce genes involved in B-cell maturation, such as OX40 and BAFF-R itself. TACI and BCMA contribute less prominently to this pathway but can amplify it in specific contexts like differentiation. Additional pathways include the PI3K/Akt axis, activated downstream of BAFF-R and TACI via association with BCR co-receptors like , where PI3K phosphorylates Akt, leading to inhibition of pro-apoptotic factors and induction of for B-cell survival. The MAPK/ERK pathway is engaged primarily by BAFF-R in an IKKα-dependent manner, promoting B-cell proliferation through ERK1/2 phosphorylation and cyclin D2 expression. In plasma cells, BCMA signaling activates via canonical induction of p62, supporting metabolic reprogramming and secretion. BAFF receptor signaling exhibits crosstalk with (BCR) and (TLR) pathways, where TACI relieves TRAF3 inhibition on SYK , synergizing with BCR to enhance activation-induced deaminase (AID) expression and class-switch recombination (CSR). This integration amplifies humoral responses without directly altering receptor binding dynamics.

Clinical significance

Involvement in diseases

Elevated levels of B-cell activating factor (BAFF) play a pathological role in various autoimmune diseases by promoting the survival and differentiation of autoreactive B cells. In systemic lupus erythematosus (SLE), serum BAFF concentrations are significantly increased and correlate positively with anti-double-stranded DNA antibody levels and disease activity, contributing to the persistence of autoreactive B cells. Similarly, in (RA), BAFF is overexpressed in the synovium, where it supports B-cell survival and local inflammation, with elevated levels detected in both serum and that associate with disease progression. In Sjögren's syndrome, BAFF expression is upregulated in inflamed salivary glands, driving B-cell infiltration and hyperactivity that exacerbate glandular damage. In immunodeficiencies, disruptions in BAFF signaling lead to impaired B-cell development and function. Mutations in the BAFF receptor (BAFF-R) gene, such as null variants, cause a block in B-cell maturation from the immature to transitional stage, resulting in profound B-cell lymphopenia and a resembling (CVID). In CVID patients, reduced BAFF-R expression on B cells has been observed, inversely correlating with disease severity and contributing to , although compensatory elevations in soluble BAFF may occur in some cases. BAFF also facilitates the survival of malignant B cells in various lymphomas through autocrine and paracrine mechanisms. In (CLL), BAFF promotes leukemic B-cell survival via interactions with receptors like BCMA and TACI, often in an autocrine loop supported by nurselike cells in the microenvironment. In , BAFF and its receptor BAFF-R are expressed on tumor cells, enhancing proliferation and resistance to . High serum BAFF levels have been linked to poor prognosis in , where it supports survival and disease progression. Beyond immune disorders, BAFF contributes to allograft rejection by activating B cells in the graft. In kidney transplant recipients, elevated pretransplantation BAFF levels predict increased risk of acute antibody-mediated rejection, correlating with donor-specific antibody development and B-cell responses against the graft. Emerging research also implicates BAFF in non-immune pathologies; in patients undergoing , elevated plasma BAFF associates with the onset of depressive symptoms, potentially through neuroinflammatory pathways.

Therapeutic modulation and developments

Belimumab, a targeting soluble BAFF, was approved by the FDA in March 2011 for the treatment of active, autoantibody-positive (SLE) in adults receiving standard therapy. In December 2020, the FDA expanded approval to include adult patients with active receiving standard therapy, marking the first new biologic for this indication in over a decade. Atacicept, a recombinant that inhibits both BAFF and APRIL by binding to TACI, demonstrated in a Phase III ORIGIN trial for , meeting its primary endpoint of reduction at week 36 in an interim analysis reported in November 2025; Vera Therapeutics subsequently submitted a Biologics License Application to the FDA via accelerated approval. Several BAFF-targeted agents remain in clinical development for autoimmune diseases. Blisibimod, a peptibody that selectively inhibits BAFF, was evaluated in Phase III trials (CHABLIS-SC1 and CHABLIS 7.5) for SLE but did not meet the primary SRI-6 endpoint, though it showed benefits in tapering and reduction. Ianalumab, a against BAFF-R, achieved statistically significant reductions in disease activity (measured by ESSDAI score) in two Phase III trials for primary Sjögren's syndrome, reported in August 2025, representing the first such success in global Phase III studies for this condition. Therapeutic modulation of BAFF primarily involves neutralization, which promotes of autoreactive and reduces circulating mature numbers by approximately 50-70% over time. For instance, treatment leads to sustained decreases in naive and activated subsets, correlating with clinical responses such as a Systemic Lupus Erythematosus Responder Index-4 (SRI-4) rate of 58% versus 44% for in pivotal trials. Challenges in BAFF inhibition include increased risk due to B-cell depletion, observed across trials with higher rates of upper respiratory and urinary tract compared to . Recent research from 2023-2025 has focused on biomarkers, with elevated pretreatment BAFF levels identified as predictors of response in SLE and immune , guiding patient selection to mitigate non-response variability.

Recombinant production

Expression systems

Recombinant production of B-cell activating factor (BAFF), also known as BLyS or TNFSF13B, relies on various expression systems to generate the soluble form, typically comprising amino acids 134-285 of the mature protein, which forms bioactive trimers essential for its TNF family structure. Bacterial systems, particularly Escherichia coli, represent the most common and cost-effective approach for high-yield production of recombinant soluble BAFF (hsBAFF). Expression is typically achieved in strains like BL21(DE3) using pET vectors under a T7 promoter, often resulting in insoluble inclusion bodies that constitute a significant portion of total cellular protein. These inclusion bodies are solubilized under denaturing conditions (e.g., 8 M urea) and refolded in vitro, often on-column during purification, to restore the native trimeric conformation and biological activity, despite the absence of glycosylation in prokaryotic hosts. Yields of up to 15 mg/L have been reported from flask cultures following refolding, with the refolded hsBAFF exhibiting full bioactivity in stimulating B-cell proliferation. To enhance expression, cDNA encoding the soluble domain is employed, along with IPTG induction at optimized temperatures to minimize aggregation. Mammalian systems, such as HEK293 and cells, are preferred for producing glycosylated hsBAFF that more closely mimics the native post-translationally modified form, which is critical for therapeutic applications requiring authentic and . Transient or stable in HEK293 cells yields soluble, N-glycosylated hsBAFF with proper trimerization, though specific production levels vary with optimization; these systems support higher fidelity folding compared to bacterial hosts. In cells, stable cell lines expressing the soluble form have achieved yields of approximately 0.55 mg/L in culture supernatant, with the protein displaying a molecular weight increase to ~20 kDa due to and confirmed B-cell stimulatory activity. These eukaryotic systems generally provide lower volumetric yields (often in the range of 0.1-1 mg/L for stable lines) but ensure functional patterns absent in bacterial expression. Other systems, including insect and yeast cells, offer alternatives for rapid or partially glycosylated production. In insect cells like , baculovirus-mediated expression enables quick generation of hsBAFF, often integrated into virus-like particles for studies, providing a versatile platform for moderate-scale production with insect-specific modifications. Yeast systems, particularly Pichia pastoris, facilitate secreted expression of N-glycosylated hsBAFF using vectors like pPIC9, yielding up to 102 mg/L from culture supernatant with partial mannose-type that increases the protein's molecular weight to ~20 while preserving trimeric structure and B-cell proliferation activity comparable to mammalian-derived forms. These non-mammalian eukaryotic options balance yield and modification needs for purposes.

Purification and applications

Purification of recombinant BAFF typically begins with affinity chromatography using Ni-NTA resin to capture His-tagged protein expressed in bacterial systems such as Escherichia coli. Following elution, the protein is often subjected to size-exclusion chromatography (SEC) on columns like Superdex 75 or 200 to isolate the biologically active trimeric form, achieving purity levels exceeding 95%. For preparations intended for in vivo applications, endotoxin removal is essential and commonly performed using polymyxin B affinity columns to reduce levels below 1 EU/μg. Yields from bacterial expression systems vary but can reach approximately 15 mg/L after refolding and purification from in flask cultures. The purified soluble trimeric BAFF exhibits good stability, remaining active for weeks when stored at in buffers containing stabilizers, with lyophilized forms stable for months at -20°C. is routinely assessed via ELISA-based assays measuring B-cell or in response to BAFF stimulation, confirming functionality at concentrations around 1-10 ng/mL. In research settings, recombinant BAFF serves as a key tool for assays evaluating B-cell maturation, survival, and , often co-stimulated with anti-IgM antibodies. It is also employed in animal models of , such as lupus-prone mice, to study BAFF's role in disease progression by administering doses that enhance B-cell responses. As a diagnostic , purified BAFF is incorporated into commercial kits for quantifying serum levels in patients with autoimmune disorders, enabling sensitive detection down to 10 pg/mL. Additionally, BAFF acts as a molecular in formulations, boosting by promoting antigen-specific production in preclinical studies against pathogens like . Key challenges in BAFF purification include preventing , which can be mitigated by incorporating non-ionic detergents like 0.01-0.1% Tween-20 during storage and steps to maintain trimer integrity. Scalability for preclinical studies requires optimization of refolding conditions and downstream processes to consistently achieve high-purity material without loss of bioactivity.

References

  1. [1]
    The BAFF/APRIL system: Emerging functions beyond B cell biology ...
    The BAFF system plays a key role in the development of autoimmunity, especially in systemic lupus erythematosus (SLE).
  2. [2]
    Current status of BAFF targeting immunotherapy in B-cell neoplasm
    Sep 2, 2024 · BAFF induces B-cell proliferation and immunoglobulin secretion, and acts as a survival factor for immature, naive, and activated B cells.
  3. [3]
    BLyS: member of the tumor necrosis factor family and B lymphocyte ...
    A member of the human TNF family, BLyS (B lymphocyte stimulator), was identified that induced B cell proliferation and immunoglobulin secretion.Missing: BAFF | Show results with:BAFF
  4. [4]
    TALL-1 is a novel member of the TNF family that is down-regulated ...
    TALL-1 is a novel member of the TNF family that is down-regulated by mitogens. J Leukoc Biol. 1999 May;65(5):680-3. Authors. H B Shu , W H Hu, H Johnson ...Missing: BAFF journal
  5. [5]
    BAFF, a novel ligand of the tumor necrosis factor family, stimulates B ...
    BAFF, a novel ligand of the tumor necrosis factor family, stimulates B cell growth ... J Exp Med. 1999 Jun 7;189(11):1747-56. doi: 10.1084/jem.189.11.1747.
  6. [6]
  7. [7]
    TNFSF13B TNF superfamily member 13b [ (human)] - NCBI
    Sep 5, 2025 · BAFF, involved in B cell activation through the NF-kappaB pathway, is related to disease activity and bone destruction in rheumatoid arthritis.Missing: exons kb
  8. [8]
    Tnfsf13b tumor necrosis factor (ligand) superfamily, member ... - NCBI
    BAFF-R expression is tightly regulated during B-cell development in mouse and human and this expression is correlated with positive selection.Missing: chromosomal | Show results with:chromosomal
  9. [9]
    Identification and characterization of a novel cytokine ... - PubMed
    THANK also strongly suppressed the growth of tumor cell lines and activated caspase-3. Although THANK had all the activities and potency of TNF, ...
  10. [10]
    TNFRSF13B - an overview | ScienceDirect Topics
    The human baff gene, encoded at 13q32-q34, contains a transmembrane domain and flanking regions in exon 1, a furin-processing site in exon 2, and a TNF homology ...
  11. [11]
    Entry - *603969 - TUMOR NECROSIS FACTOR LIGAND ... - OMIM
    (2005) analyzed synovial tissue specimens from 72 patients with rheumatoid arthritis for TNFSF13 and TNFSF13B production and TNFSF13/TNFSF13B receptor ...
  12. [12]
    Induction of BAFF expression by IFN‐γ via JAK/STAT signaling ...
    Dec 27, 2012 · Collectively, these results suggest that IFN-γ induces BAFF expression in human intestinal epithelial cells through JAK/STAT signaling pathways ...Ifn-γ Induces Production Of... · Jak/stat Signaling Pathway... · Ifn-γ Induces The...
  13. [13]
    Tissue expression of TNFSF13B - Summary - The Human Protein Atlas
    The RNA data was used to cluster genes according to their expression across tissues. Clusters contain genes that have similar expression patterns, and each ...Missing: intronic enhancers
  14. [14]
    [PDF] Recombinant Human BAFF/BLyS/TNFSF13B His-tag
    Mature human BAFF consists of a 46 amino acid (aa) cytoplasmic ... Within aa 134-285 of the ECD, human BAFF shares 72% aa sequence identity with mouse BAFF.
  15. [15]
    Predicted aa sequences of pig, human and mouse BAFF. The ...
    ... BAFF (86% and 69% amino acid (aa) identity, respectively). For this domain, homology of porcine to human BAFF is higher than homology of murine to human BAFF ..
  16. [16]
    Analysis of TNFSF13B polymorphisms and BAFF expression in ...
    Apr 12, 2022 · The TNFSF13B gene encodes for BAFF protein and has been mapped to chromosome 13q32-34 (Schneider et al., 1999). Three single nucleotide ...<|control11|><|separator|>
  17. [17]
    Analysis of TNFSF13B polymorphisms and BAFF expression in ...
    Apr 12, 2022 · The rs9514828 (-871 C > T) polymorphism was associated with increased gene expression in RA patients. Also, sBAFF levels were higher in both ADs ...
  18. [18]
  19. [19]
    Baff Mediates Survival of Peripheral Immature B Lymphocytes
    BAFF treatment allowed T2 B cells to survive and differentiate into mature B cells in response to signals through the BCR. The T2 and the marginal zone (MZ) B ...
  20. [20]
    BAFF and BAFF-Receptor in B Cell Selection and Survival - Frontiers
    The BAFF-receptor (BAFFR) is encoded by the TNFRSF13C gene and is one of the main pro-survival receptors in B cells.Abstract · BAFFR and B Cell Survival · BAFFR Deficiency in Humans
  21. [21]
    Crucial Role for BAFF-BAFF-R Signaling in the Survival and ...
    (1999) Mice transgenic for BAFF develop lymphocytic disorders along with autoimmune manifestations. J Exp Med 190: 1697–1710. 15. Khare SD,Sarosi I,Xia XZ ...
  22. [22]
    Innate control of B cell responses - PMC - NIH
    Innate Ig-inducing signaling pathways. TACI and TLRs. BAFF and APRIL trigger CD40-independent IgM production, CSR and plasma cell survival and differentiation ...
  23. [23]
    The Interplay of IL-21 and BAFF in the Formation and ... - Frontiers
    In combination with CD40 ligation, IL-21 induces CSR, plasma cell differentiation, and Ig production from B cells isolated from various lymphoid compartments ...
  24. [24]
    BAFF and the plasticity of peripheral B cell tolerance - PMC - NIH
    BAFF and its receptors play a crucial role in peripheral B cell selection and survival, by dictating the set point for mature primary B cell numbers.Introduction · B Cells Undergo Both... · Baff/baffr Signaling...Missing: original | Show results with:original
  25. [25]
    BAFF and selection of autoreactive B cells - PMC - PubMed Central
    In this review, we focus on the evidence supporting a role for BAFF, and its homologue APRIL, in regulating the selection and survival of autoreactive B cells.Missing: clones | Show results with:clones
  26. [26]
    Targeting BAFF in autoimmunity - PMC - NIH
    Murine BAFF-receptor residues 168–175 are essential for optimal CD21/35 expression but dispensable for B cell survival. Mol Immunol. 2009;47:590–599. doi ...Missing: ratio | Show results with:ratio
  27. [27]
    BAFF: a local and systemic target in autoimmune diseases - PMC
    BAFF (B lymphocyte activating factor of the tumour necrosis factor family) is a vital homeostatic cytokine for B cells that helps regulate both innate and ...Missing: definition | Show results with:definition
  28. [28]
    The function of BAFF on T helper cells in autoimmunity - PMC
    B cell-activating factor belonging to the TNF family (BAFF) exerts its pathogenic role in supporting the survival and proliferation of B cells, regulating class ...Missing: definition | Show results with:definition
  29. [29]
    BAFF knockout improves systemic inflammation via regulating ...
    Jan 16, 2015 · Our group is the first to report that B-cell-activating factor (BAFF) is produced from adipocytes and functions as a proinflammatory adipokine.
  30. [30]
    B-Cell-Activating Factor Depletion Ameliorates Aging-Dependent ...
    Jul 20, 2020 · In this study, we show that systemic BAFF depletion improves aging-dependent insulin resistance. In aged (10-month-old) BAFF −/− mice, glucose tolerance and ...
  31. [31]
    Depletion of B cell-activating factor attenuates hepatic fat ... - Nature
    Jan 30, 2019 · Our data revealed that BAFF serves as a potential stimulator of unhealthy adipose-tissue expansion by triggering inflammation and fibrosis.
  32. [32]
  33. [33]
  34. [34]
  35. [35]
  36. [36]
  37. [37]
  38. [38]
  39. [39]
    A New Era for the Treatment of Lupus
    May 22, 2023 · Belimumab was initially approved in March of 2011 for the treatment of lupus (after decades without new FDA-approved drugs), and in December of ...
  40. [40]
    [PDF] 2022 Benlysta (belimumab) - FDA
    Jun 9, 2022 · On December 16, 2020, belimumab was approved for the treatment of adult patients with active lupus nephritis who are receiving standard therapy.
  41. [41]
  42. [42]
    Phase III trial results with blisibimod, a selective inhibitor of B-cell ...
    Mar 21, 2018 · Although the SRI-6 end point was not met, blisibimod was associated with successful steroid reduction, decreased proteinuria and biomarker responses.
  43. [43]
    Novartis ianalumab first drug to reduce disease activity and patient ...
    Oct 29, 2025 · Novartis ianalumab first drug to reduce disease activity and patient burden in Sjögren's disease Phase III trials. Oct 29, 2025. NEPTUNUS-1 ...
  44. [44]
    Reductions in circulating B cell subsets and immunoglobulin G ...
    Feb 7, 2022 · Belimumab treatment up to 312 weeks (6 years) resulted in substantial decreases in several circulating B cell subsets and IgG levels.
  45. [45]
    BENLYSTA (belimumab) for Lupus and Lupus Nephritis | for HCPs
    BENLYSTA was proven to reduce lupus symptoms in patients with lupus. Up to 61% of patients had reduced disease activity (SRI-4) at Week 52.About Lupus Nephritis · Efficacy for Lupus Nephritis · About Lupus · Vial Calculator
  46. [46]
    BAFF- and APRIL-targeted therapy in systemic autoimmune diseases
    BAFF, also called B lymphocyte stimulator (BLyS), is a B cell activation factor which is mainly expressed by monocytes, macrophages, and activated T cells. BAFF ...
  47. [47]
    BAFF as a predictive marker for treatment response in immune
    This work aimed to study serum BAFF levels in the Egyptian ITP patients, and the relation between its level and response to steroid therapy.Missing: Challenges | Show results with:Challenges
  48. [48]
    What BAFF inhibitors are in clinical trials currently? - Patsnap Synapse
    Mar 11, 2025 · One major challenge is the variability in the immune response among patients. Since BAFF is fundamentally involved in normal immune homeostasis ...
  49. [49]
  50. [50]
    Recombinant Human BAFF/BLyS/TNFSF13B Protein, 10056-HNCH
    Recombinant Human BAFF/BLyS/TNFSF13B Protein (Ala134-Leu285) ECD is expressed in HEK293 Cells. Purity is > 95 % as determined by SDS-PAGE.
  51. [51]
  52. [52]
    Preparation of the recombinant BAFF. - ResearchGate
    Purification was sequentially performed by the anion exchanger and Ni 2+ affinity column. Analyzed by SDS-PAGE, smBAFF and pNO 2 Phe 65 smBAFF have an ...
  53. [53]
    Expression, purification, and characterization of recombinant human ...
    Herein, the cDNA coding for the extracellular domain of the BAFF (hsBAFF) has been cloned into the secreting expression organism Pichia pastoris.
  54. [54]
    ToxinEraser™ Endotoxin Removal Kit - GenScript
    30-day returnsIt is based on affinity matrix of modified polymyxin B (PMB), which allows highly efficient endotoxin removal. The final endotoxin level can be decreased to ...Missing: BAFF | Show results with:BAFF
  55. [55]
    Expression, refolding, and characterization of human soluble BAFF ...
    Here, a recombinant form of the extracellular domain of the BAFF (hsBAFF) was expressed in Escherichia coli BL21(DE3) under the control of a T7 promoter.Missing: systems | Show results with:systems
  56. [56]
    BAFF Protein Human Recombinant | TNFSF13B Antigen - Prospec Bio
    3–4 day deliveryStability. Lyophilized BAFF although stable at room temperature for 3 weeks, should be stored desiccated below -18°C. Upon reconstitution BAFF should be ...Missing: yield | Show results with:yield
  57. [57]
    Recombinant Human BAFF carrier-free - BioLegend
    Aug 27, 2025 · Recombinant human BAFF induces the proliferation of mouse B cells (stimulated with anti-IgM) in a dose dependent manner.
  58. [58]
    Neutralizing B-Cell–Activating Factor Antibody Improves Survival ...
    Functional studies show that recombinant BAFF protects MM cells against dexamethasone-induced apoptosis accompanied by an increase in survival proteins ...
  59. [59]
    Human BAFF/BLyS/TNFSF13B DuoSet ELISA - R&D Systems
    Rating 4.0 (1) · $45 deliveryThis DuoSet ELISA Development kit contains the basic components required for the development of sandwich ELISAs to measure natural and recombinant human BAFF / ...
  60. [60]
    Expression of B-Cell Activating Factor Enhances Protective Immunity ...
    These results show that BAFF enhances antigen-specific humoral immunity and suggest that BAFF is a promising cytokine for the development of an improved ...
  61. [61]
    The Role of Tween 80 in Protein Solubilization and Stabilization
    Mar 10, 2020 · Tween 80 is essential to protein solubilization and stabilization and protects protein from agitation-induced aggregation.