Fact-checked by Grok 2 weeks ago

Bafilomycin

Bafilomycins are a family of plecomacrolide macrolide antibiotics produced by various species of Streptomyces bacteria, with bafilomycin A1 being the most extensively studied and prototypical member. First isolated in 1983 from Streptomyces griseus subsp. sulphurus (strain TÜ 1922), these compounds are characterized by a complex 16-membered macrocyclic lactone ring incorporating conjugated diene systems and a hemiketal functionality, conferring their unique biological activity.42898-4/fulltext) Bafilomycin A1 specifically and potently inhibits vacuolar-type H⁺-ATPases (V-ATPases), multi-subunit proton pumps that acidify intracellular compartments such as lysosomes, endosomes, and Golgi-derived vesicles, thereby disrupting pH-dependent cellular processes including autophagy, endocytosis, and protein degradation. This V-ATPase inhibition by bafilomycin A1 prevents lysosomal acidification, which blocks the fusion of autophagosomes with lysosomes and leads to the accumulation of autophagic vacuoles, making it a widely used pharmacological tool to study autophagic flux in mammalian cells. Beyond autophagy, bafilomycins exhibit antifungal, antiproliferative, and osteoclast-inhibitory effects; for instance, they suppress bone resorption by inhibiting V-ATPase in osteoclasts, highlighting potential therapeutic relevance in conditions like osteoporosis.42898-4/fulltext) However, their clinical development has been hampered by significant cytotoxicity, including induction of apoptosis and disruption of lysosomal cholesterol trafficking, which limits their use primarily to in vitro and ex vivo research applications. Ongoing studies explore bafilomycin derivatives and biosynthetic engineering in Streptomyces to mitigate toxicity while preserving efficacy, aiming to expand their utility in cancer therapy and infectious disease research.

Discovery and Biosynthesis

Initial Discovery

Bafilomycin A1 was first isolated in 1983 from the fermentation broth of the actinomycete griseus subsp. sulphurus strain TU 1922 during a screening program for novel microbial metabolites at the in , led by Gerhard Werner and Hanspaul Hagenmaier. The isolation process involved extraction of the culture broth followed by purification via , yielding several related compounds including bafilomycins A1, A2, B1, B2, C1, and C2. Initial biological evaluations demonstrated antifungal activity against fungi such as Mucor miehei via disc diffusion assays (inhibition zones of 23–39 mm), alongside moderate effects on . These findings positioned the bafilomycins as promising candidates in early antibiotic efforts targeting fungal pathogens. The of bafilomycin A1 was elucidated shortly thereafter through a combination of (NMR) spectroscopy and , revealing a novel 16-membered core characteristic of the plecomacrolide family. The naming convention "bafilomycin" derives from the producing strain, reflecting its microbial origin. Subsequent studies identified its mechanism as inhibition of vacuolar-type H⁺-ATPase (), though this was not recognized at the time of .

Producing Organisms

Bafilomycins are primarily produced by actinomycetes, a group of Gram-positive, filamentous bacteria predominantly found in soil environments. Key producers include species of Streptomyces such as S. griseus and S. hygroscopicus, as well as members of the genus Kitasatospora, including K. setae, K. griseola, and K. cheerisanensis. These organisms were first identified as bafilomycin sources through isolation from soil samples, with S. griseus serving as the initial producer of bafilomycin A1 in early discoveries (https://www.sigmaaldrich.com/US/en/product/sigma/b1793). The ecological context of these producers centers on their role in soil microbial communities, where they contribute to nutrient cycling and secondary metabolite production as a defense mechanism against competitors (https://bjbas.springeropen.com/articles/10.1186/s43088-025-00622-0). Isolation of bafilomycin-producing actinomycetes typically involves collecting soil from diverse habitats, such as terrestrial or marine-adjacent sediments, followed by pretreatment and culturing on selective media like starch-casein or ISP media to suppress faster-growing and promote sporulating filaments (https://www.nature.com/articles/s41598-025-09357-5). This approach has yielded strains from high-altitude soils and deep-sea-derived sediments, highlighting the adaptability of these actinomycetes to varied geochemical conditions (https://pubs.acs.org/doi/abs/10.1021/np900632r). Natural habitats rich in , such as or agricultural soils, favor their growth, with isolation techniques often incorporating air-drying or heat pretreatment to enrich for resilient spores (https://microbiologyjournal.org/actinomycetes-isolation-cultivation-and-its-active-biomolecules/). Advancements in genomics during the 2010s enabled the identification of bafilomycin biosynthetic gene clusters (BGCs) through whole-genome sequencing of producer strains. For example, the 87.4 kb BGC in S. griseus DSM 2608 was fully sequenced in 2013, revealing modular polyketide synthase genes essential for macrolide assembly (https://pmc.ncbi.nlm.nih.gov/articles/PMC3679948/). Similarly, BGCs in K. setae KM-6054 and K. cheerisanensis KCTC 2395 were characterized around 2014–2017, spanning 70–80 kb and containing multiple open reading frames for precursor biosynthesis and tailoring (https://www.nature.com/articles/ja201733) (https://link.springer.com/article/10.1007/s12275-015-4340-0). At least six such BGCs have been reported across Streptomyces and Kitasatospora species, facilitating comparative analyses of evolutionary conservation (https://pmc.ncbi.nlm.nih.gov/articles/PMC7827423/). Recent genomic mining has identified additional producers, such as deep-sea-derived Streptomyces samsunensis OUCT16-12 yielding novel antiproliferative bafilomycins (as of 2023), and a 2025 review documents over 50 natural bafilomycin derivatives from actinomycetes up to 2024. Strain variations significantly influence bafilomycin yields, with wild-type isolates exhibiting production ranging from trace amounts to milligrams per liter depending on media and environmental cues. Genetic engineering has addressed low yields in natural strains; for instance, overexpression of regulatory genes like orf1 and bafG in Streptomyces lohii increased bafilomycin A1 production by up to 10-fold through enhanced pathway activation (https://pmc.ncbi.nlm.nih.gov/articles/PMC7827423/). Such modifications in engineered Streptomyces hosts demonstrate potential for scalable biosynthesis while preserving the organisms' ecological relevance in natural product discovery (https://www.mdpi.com/1660-3397/19/1/29).

Biosynthetic Pathway

The biosynthetic pathway of bafilomycin is mediated by a modular type I (PKS) system in species, such as Streptomyces lohii and Kitasatospora setae, which assembles the macrolactone core through iterative chain elongation and subsequent tailoring steps. The PKS consists of five large open reading frames (bafA I–V) spanning approximately 59 kb, encoding 11 extension plus a loading module that incorporate extender units to build the chain. Chain elongation begins with the loading module accepting a propionyl-CoA starter unit, followed by 11 cycles of using primarily methylmalonyl-CoA extenders (in modules 1, 3, 4, 7–10) and (in modules 2 and 6), with modules 5 and 11 incorporating methoxymalonyl-CoA for specific C-methyl and methoxy functionalities. Each contains catalytic domains including ketosynthase (KS), acyltransferase (AT), and (ACP), with additional domains like ketoreductase (KR), (DH), and enoylreductase (ER) that shape the intermediates; for instance, DH domains in modules 6–8 and 10–11 facilitate to form conjugated systems essential for the molecule's . The full chain is released and cyclized by a thioesterase domain in the final to form the 16-membered macrolactone ring. Key enzymes include BafA, which encompasses the KS domains across all modules for decarboxylative condensation during elongation, and BafD, an enoyl reductase in module 8 that reduces the α,β-unsaturated carbonyl to saturate specific positions in the chain. Post-PKS modifications further diversify the scaffold, such as the attachment of a C5N (5-aminolevulinic acid-derived) moiety via by BafY and activation by BafX, or fumarate addition to hydroxyl groups mediated by Orf2 and Orf3, though these steps occur after core assembly. Regulation of the pathway involves pathway-specific activators like BafG, a homolog of the pleiotropic transcriptional regulator AfsR that enhances expression of the , and Orf1, a LuxR-family protein responsive to quorum-sensing signals. In streptomycetes, environmental factors such as can influence production generally, with optimal bafilomycin yields observed at neutral around 7.0–7.1 in culture media.

Chemical Structure and Properties

Molecular Structure

Bafilomycin A1 features a core 16-membered ring system, which serves as the defining scaffold for the bafilomycin family of antibiotics. This ring incorporates two conjugated units that contribute to its extended conjugation and rigidity, along with a hemiketal moiety that links the macrocycle to a deoxysugar . The overall is typical of plecomacrolide antibiotics, with the formed between a carboxylic acid-derived carbonyl at one end and an at the opposite position of the ring. Key functional groups on the include an bridging carbons and C14, which imparts strain and reactivity, as well as hydroxyl groups at and that influence hydrogen bonding and . Additionally, a is present at C21, contributing to the molecule's and potentially its binding interactions. These elements are arranged in a polypropionate-derived that folds into the macrocycle during . The molecular formula of bafilomycin A1 is C35H58O9, corresponding to a molecular weight of approximately 622 . The of bafilomycin A1 encompasses 12 chiral centers, with configurations established through detailed spectroscopic analysis, including NMR, and later confirmed by in the 1980s. This complex array of stereocenters ensures the precise three-dimensional shape required for its , with the adopting a specific conformation stabilized by intramolecular hydrogen bonds involving the hemiketal and hydroxyl groups.

Physicochemical Properties

Bafilomycin A1 exhibits poor solubility in , typically described as insoluble or with a maximum solubility of approximately 0.0155 mg/mL, which limits its use in aqueous formulations. In contrast, it demonstrates good solubility in organic solvents, reaching up to 5 mg/mL in DMSO and , and similar levels in , facilitating its dissolution for experimental applications. The compound is sensitive to light, particularly in solution, necessitating protection from prolonged exposure during handling and storage to prevent degradation. Bafilomycin A1 is also thermally labile, with recommendations to store it at -20°C under desiccated conditions to ensure long-term stability, as exposure to higher temperatures can compromise its integrity. Its lipophilicity is reflected in a logP value of approximately 4.5 (ranging from 3.89 to 5.08 across predictive models), contributing to effective membrane permeability in biological systems. Spectroscopically, bafilomycin A1 shows characteristic UV absorption maxima at 248 nm and 288 nm, arising from its conjugated diene moieties in the macrolide structure, which enables reliable quantification via UV detection in chromatographic assays.

Structural Variants

The bafilomycin family encompasses a series of structurally related plecomacrolide antibiotics, primarily distinguished by modifications to the side chain attached to the core 16-membered macrolactone ring. Bafilomycin A1 serves as the prototype, featuring a hydroxyl group at C-21 and a methoxy substituent at C-23 in its side chain, which confers high potency as a V-ATPase inhibitor with an IC50 in the low nanomolar range (approximately 10-100 nM depending on the assay system). In contrast, bafilomycin A2 is a desmethyl variant lacking the C-21 methoxy group (replaced by a hydroxyl), resulting in slightly reduced inhibitory activity against V-ATPase, with IC50 values typically 2-5 times higher than A1 due to altered binding affinity. The B and C subfamilies exhibit further side chain alterations that impact potency. Bafilomycin B1 features a flavensomycinyl-like extension at C-21 and lacks the C-21 methoxy present in , leading to a side chain with two hydroxyl groups (-CH2-CH(OH)-CH(OH)-iPr) and diminished inhibition ( ~200-500 nM), approximately 5-10-fold less potent than . Similarly, bafilomycin C1 incorporates a fumarate moiety at C-21 instead of the hydroxyl or methoxy, further reducing activity ( ~1-5 μM), while B2 and C2 variants include a methoxy at C-19 in addition to these changes, partially restoring potency in some assays but still inferior to the A series. Bafilomycin D is characterized by opening of the ring and introduction of a carbonyl at C-19, showing the lowest potency among early variants ( >10 μM) and has been isolated from marine species, where it demonstrates enhanced compared to under physiological conditions. Structure-activity relationship studies highlight key motifs for inhibition within the family. The functionality in the macrolactone core is essential for high-affinity binding to the enzyme's c-subunit, as its absence or modification abolishes activity. Modifications to the conjugated system (spanning Δ2,3, Δ4,5, Δ10,11, and Δ12,13) influence selectivity; for instance, or extension of the units reduces specificity for mammalian V-ATPases while potentially enhancing activity against microbial targets. The C-7 hydroxyl group also plays a critical role in stabilizing interactions with the target, and variations primarily modulate potency rather than . Post-2020 isolates from marine have expanded the family with structurally novel variants exhibiting improved stability or altered bioactivity profiles. For example, bafilomycins P and Q, featuring a ring system via and cyclization at the , display enhanced thermal stability and ( 1-5 μM against lines). Other recent analogs, such as ring-opened bafilomycins R, S, and T from deep-sea sediments, incorporate modifications to the for better selectivity in antiproliferative applications. These variants underscore the biosynthetic plasticity of marine actinomycetes in generating diversified structures.

Target and Mechanism of Action

V-ATPase Structure and Function

The vacuolar H⁺-ATPase () is a multi-subunit complex that functions as an ATP-driven , essential for acidifying intracellular compartments and maintaining cellular . It consists of two principal domains: the membrane-embedded V₀ domain, which forms the proton-translocating pore, and the cytosolic V₁ domain, responsible for . The V₀ domain integrates into lipid bilayers to facilitate proton movement across membranes, while the V₁ domain protrudes into the , harnessing from ATP to drive this process. This architecture enables to generate electrochemical proton gradients against concentration differences, powering secondary transport and enzymatic activities in various cellular contexts. Eukaryotic V-ATPases are composed of 14 distinct subunit types, assembled into a highly organized . The V₁ domain includes three copies each of the catalytic subunits A and B (A₃B₃), which form a hexameric head responsible for ATP binding and hydrolysis, along with regulatory subunits C, D, E, F, G, and H. In the V₀ domain, the a-subunit serves as a that coordinates proton translocation, interacting with a ring of multiple c-subunits (including c, c', and c'') to form the proton-conducting pathway; additional subunits such as d and e stabilize the integration. These subunits exhibit tissue-specific isoforms, allowing functional adaptation, with the a-subunit isoforms particularly influencing targeting and activity. The enzyme operates through a rotary mechanism, where in the V₁ domain induces conformational changes that a central stalk and the c-ring within V₀. This , occurring in 120° steps per ATP molecule hydrolyzed, drives protons from the into the lumenal side via essential glutamate residues on the c-subunits, which become protonated and deprotonated sequentially through interaction with the a-subunit's hemichannels. The process establishes a proton motive , with the typically yielding three to four protons translocated per ATP consumed, enabling efficient acidification against steep gradients. V-ATPases are localized to various membranes, including those of lysosomes, endosomes, and the Golgi apparatus, where they regulate the acidic (around 4.5–6.0) necessary for degradative enzymes and receptor recycling. In specialized cells, such as osteoclasts and renal intercalated cells, V-ATPases reside on the plasma membrane to extrude protons extracellularly, supporting and urinary acidification, respectively. This localization ensures compartmental pH control critical for cellular trafficking, nutrient uptake, and signaling.

Bafilomycin Binding and Inhibition

Bafilomycins bind to the V0 domain of vacuolar H+-ATPase (V-ATPase) within a hydrophobic pocket located on the cytosolic side of the membrane-spanning c-subunit. This pocket is formed primarily by transmembrane helices 1, 2, and 4 of the c-subunit, where the macrolide ring and conjugated tetraene chain of bafilomycin A1 engage in van der Waals interactions with hydrophobic residues such as Met-53, Ile-56, Val-60, and Ile-67 from one c-subunit, and Leu-133, Phe-137, and Val-140 from the adjacent c-subunit. The epoxide group at the tetraene terminus contributes to the snug fit within this pocket, enhancing binding affinity through additional hydrophobic contacts, while the 7'-hydroxyl group forms a hydrogen bond with Tyr-144. Cryo-electron microscopy (cryo-EM) structures confirm that up to six bafilomycin A1 molecules can bind the c-ring, with each primarily associating with two adjacent c-subunits, stabilizing the complex through these interactions. The inhibition by bafilomycin is non-competitive and allosteric, targeting the proton-translocating V0 sector without directly interfering with at the sector. By occupying the hydrophobic pocket, bafilomycin A1 sterically hinders the rotational movement of the c-ring, which is essential for proton translocation across the membrane during function. This blockade occurs independently of the enzyme's , as evidenced by preserved rates in inhibited complexes, while proton pumping is abolished. The potency of bafilomycin A1 is high, with values typically ranging from 0.4 to 100 nM depending on the and source , reflecting its nanomolar efficacy in eukaryotic systems. Structural insights from cryo-EM studies since have elucidated the mechanistic basis of this inhibition, revealing how bafilomycin induces a conformational lock on the c-ring. High-resolution structures of bafilomycin A1-bound (at ~3.2–3.6 ) show that the inhibitor disrupts key interfaces between the c-ring and the stator subunit a, preventing the essential counterclockwise rotation driven by V1 . This allosteric constraint specifically impairs proton conduction through the V0 channel without altering V1-V0 association or rotational substeps. Bafilomycins exhibit high selectivity for eukaryotic V-ATPases over bacterial F-ATPases and mitochondrial F-type ATPases, owing to conserved binding residues in the eukaryotic c-subunit that are absent or divergent in prokaryotic and organellar counterparts. This preference is attributed to structural differences in the proteolipid ring and proton pathway, with bafilomycin showing no inhibitory effect on F-ATPase activity even at micromolar concentrations. Such selectivity underscores its utility as a tool for targeting vacuolar acidification in eukaryotic cells.

Cellular and Physiological Effects

Autophagy Inhibition

Bafilomycin inhibits by targeting the vacuolar H+-ATPase (), which is essential for lysosomal acidification, thereby blocking the maturation of autolysosomes and trapping autophagosomes within the . This disruption prevents the of autophagosomes with lysosomes and the subsequent of their contents, leading to the accumulation of lipidated LC3-II, a hallmark marker of autophagosome buildup. The blockade specifically occurs at the late stage of autophagic flux, halting the process after autophagosome formation without interfering with earlier vesicular trafficking steps. In autophagy research, bafilomycin serves as the gold standard inhibitor for assessing autophagic flux through methods such as Western blot analysis of LC3-II levels, where its addition causes a detectable increase in LC3-II if flux is active. Typical experimental protocols employ concentrations of 100-400 nM for 4-24 hours to effectively inhibit lysosomal function while minimizing cytotoxicity. This approach allows researchers to distinguish between increased autophagosome formation and impaired degradation, providing a reliable measure of autophagic activity across various cell types and conditions. Bafilomycin exhibits minimal direct effects on upstream autophagy regulators, such as signaling or the initiation of biogenesis, ensuring its primary action remains confined to lysosomal disruption. Recent studies have highlighted its role in blockade contributing to antibody-dependent cellular cytotoxicity (ADCC) resistance in cancer cells, where inhibition alters cellular responses to immune-mediated killing without affecting early autophagic induction.

Apoptosis Induction

Bafilomycin, a selective of the vacuolar H+-ATPase (), disrupts lysosomal acidification, leading to lysosomal membrane permeabilization (LMP) that releases cathepsins into the . These lysosomal s, particularly and D, then trigger the activation of pro-apoptotic proteins Bax and Bak by promoting their oligomerization on the mitochondrial outer membrane. This process initiates the intrinsic apoptotic pathway, distinct from direct mitochondrial damage, and is amplified in cells with elevated lysosomal activity. The released cathepsins contribute to the activation of stress-activated protein kinases, including JNK and p38 MAPK, which phosphorylate downstream targets to enhance apoptotic signaling. At concentrations of 50-200 nM, bafilomycin promotes the cleavage and activation of initiator caspase-9 and effector caspase-3, resulting in poly(ADP-ribose) polymerase (PARP) cleavage and executioner phase of apoptosis. This caspase-dependent mechanism is modulated by upstream autophagy inhibition, which accumulates damaged organelles and sensitizes cells to death signals. Apoptosis induction by bafilomycin exhibits cell-type specificity, with pronounced effects in cancer cells exhibiting high lysosomal activity, such as those in colon, hepatocellular, and pancreatic carcinomas, where overexpression supports survival under stress. The response is time-dependent, with peak apoptotic markers—including Bax translocation, activation, and cell viability loss—observed at 24-48 hours post-treatment. Recent studies highlight bafilomycin's role in colon cancer, where low-dose treatment (1.5-2 nM) impairs endolysosomal iron homeostasis, indirectly disrupting mitochondrial function through lysosomal damage and contributing to . This mechanism underscores potential therapeutic synergy in colorectal malignancies, where lysosomal dysfunction exacerbates mitochondrial stress and caspase-independent .

Ion Transport Disruption

Bafilomycin, by inhibiting the vacuolar H⁺-ATPase () , disrupts the electrochemical gradients essential for maintaining lysosomal and endosomal homeostasis, extending its effects beyond proton transport to other cations. Inhibition of indirectly modulates K⁺ channels through alterations in lysosomal , promoting a K⁺ leak that contributes to hyperpolarization and impairs cellular volume regulation. This occurs as the collapse of the proton gradient reduces the counterion flux typically balanced by K⁺ influx, leading to passive K⁺ efflux via leak conductances such as TMEM175, an K⁺ channel that stabilizes lysosomal and function. In parallel, bafilomycin's independent K⁺ activity facilitates K⁺ transport across membranes, exacerbating imbalances in lysosomes and mitochondria. Bafilomycin induces Ca²⁺ dysregulation by enhancing endoplasmic reticulum (ER) Ca²⁺-ATPase () activity, as demonstrated in recent studies on rat liver and human colorectal tissues. Specifically, exposure to bafilomycin increases activity by up to threefold in ER fractions, driven by Ca²⁺ release from acidic stores like lysosomes following inhibition; this creates localized Ca²⁺ "hot spots" near the ER, activating to overload the ER with Ca²⁺ and trigger Ca²⁺-induced Ca²⁺ release (CICR) channels, resulting in cytosolic Ca²⁺ spikes. These spikes disrupt overall Ca²⁺ signaling and in affected cells. Crosstalk between V-ATPase inhibition and Na⁺/H⁺ exchangers (NHEs) leads to compensatory activity of NHEs in endosomal and lysosomal compartments, attempting to restore pH balance amid alkalinization. For instance, in endocytic vesicles, NHE-mediated Na⁺ influx coupled with H⁺ efflux partially offsets the reduced acidification from blockade. Studies combining bafilomycin with NHE inhibitors demonstrate further alkalinization of endosomal pH, indicating the compensatory role of NHEs. This compensatory mechanism helps mitigate excessive alkalinization but can further perturb Na⁺ gradients in acidified organelles. At high doses exceeding 1 μM, these ion transport disruptions culminate in physiological impacts such as cellular swelling and , primarily through osmotic imbalances and dysfunction. Mitochondrial swelling arises from K⁺-driven influx facilitated by bafilomycin's properties, while lysosomal ion deregulation contributes to overall cell volume dysregulation and permeabilization, triggering caspase-independent necrotic pathways characterized by rupture.

Research and Therapeutic Applications

Anticancer Effects

Bafilomycin A1 exerts anticancer effects primarily through inhibition of , which disrupts the acidic (TME) essential for cancer cell survival under . In hypoxic tumors, cancer cells rely on for energy, producing that acidifies the and promotes and to ; bafilomycin A1 counters this by blocking V-ATPase-mediated proton , thereby increasing extracellular pH and inhibiting glycolysis-dependent . This pH modulation has been shown to suppress tumor growth and in vivo in GH3 xenografts, with a 30% reduction in observed alongside elevated extracellular pH measured via 31P-MRS. Synergistic interactions further enhance bafilomycin's therapeutic potential in . By impairing endosomal and lysosomal acidification, bafilomycin A1 prevents the and of chemotherapeutic agents like doxorubicin, increasing their cytosolic accumulation and cytotoxicity in hepatic cancer cells such as HepG2. This endosomal trapping mechanism potentiates doxorubicin-induced and lysosomal membrane permeabilization. Additionally, in gliomas, bafilomycin A1 overcomes drug resistance by inhibiting , sensitizing cells to Src inhibitors like Si306 and enhancing overall anti-tumor efficacy in combination therapies. Preclinical studies demonstrate potent activity against various cancer types, with values typically in the low nanomolar range. In cell lines like MDA-MB-231 and SK-BR-3, bafilomycin A1 inhibits at 5–20 nM, often in with agents like epirubicin. For colon cancer, effective concentrations of 1.5–2 nM induce caspase-independent in lines such as LoVo and SW480 via endolysosomal dysfunction and iron disruption, sparing normal colon fibroblasts (e.g., CCD-18Co) at these doses, as reported in 2025 investigations. These effects are partly mediated by brief inhibition of and induction of , contributing to reduced tumor viability without excessive off-target impact on non-malignant cells. Despite promising preclinical results, bafilomycin's clinical translation is hindered by systemic that restricts dosing to low levels (e.g., 0.1–10 mg/kg in mice, where higher doses cause adverse effects). As of 2025, no clinical trials have evaluated bafilomycin A1 for anticancer applications, primarily due to its narrow therapeutic window and potential for off-target inhibition in normal tissues.

Antimicrobial Effects

Bafilomycin, a specific of vacuolar-type H⁺-ATPase (), exhibits activity by blocking vacuolar acidification, a process conserved across eukaryotic pathogens including fungi. In , particularly azole-resistant strains, bafilomycin A1 synergizes with azole such as and , enhancing their efficacy through combined disruption of pH-dependent cellular processes and inhibition. This results in fractional inhibitory concentration indices as low as 0.25, indicating potent cooperative effects that restore in resistant isolates. Similarly, in , bafilomycin B1 treatment impairs vacuolar acidification, leading to defects in biosynthesis, sporulation, and overall hyphal growth, as evidenced by morphological changes akin to V-ATPase subunit mutants. These effects underscore bafilomycin's role in targeting fungal , with observed growth inhibition at concentrations around 1-5 μM . In antiparasitic applications, bafilomycin disrupts the digestive (food vacuole) acidification in , the causative agent of , by inhibiting activity on the vacuolar membrane. This deacidification, achieved at concentrations as low as 61 nM, impairs digestion and detoxification, causing vacuole swelling, accumulation of undigested material, and parasite death within hours. Studies in erythrocyte-stage parasites demonstrate fractional inhibitory concentrations in the nanomolar to low micromolar range (1-10 μM), with heightened sensitivity in V-ATPase subunit-deficient strains, highlighting its potential as an adjunct to existing antimalarials like by exacerbating dysregulation essential for parasite nutrient uptake. Bafilomycin demonstrates antiviral effects primarily against enveloped viruses reliant on endosomal acidification for entry, such as A and B viruses. By preventing V-ATPase-mediated proton pumping, bafilomycin A1 neutralizes endosomal at concentrations of 0.1 μM, blocking the low-pH trigger for conformational changes necessary for viral uncoating and genome release in host cells like MDCK. This inhibition is most effective when applied within 5-10 minutes post-infection, reducing by over 90% without affecting later replication stages, and is reversible upon removal as acidified compartments reform. Recent investigations have also revealed bafilomycin's modulation of (ADCC), where at 500 nM it nearly abolishes NK cell-mediated killing of cells by altering immune signaling and surface receptor dynamics, as shown in 2025 models. Antibacterial activity of bafilomycin is limited but notable against intracellular pathogens like , where it targets host and bacterial to disrupt phagolysosome maturation. In models, bafilomycin A1 inhibits intracellular mycobacterial growth with an effective concentration in the low nanomolar range (e.g., 10–100 nM), amplifying host cell cytotoxicity and promoting late / in infected cells by countering the pathogen's PtpA-mediated blockade of phagosomal acidification. This effect is dependent on bacterial activity, as it is abolished in PtpA knockouts, positioning bafilomycin as a host-directed that enhances lysosomal killing of persistent intracellular without broad-spectrum extracellular activity.

Neurodegenerative Disease Applications

Bafilomycin A1 has been instrumental in elucidating the role of lysosomal degradation in clearing alpha-synuclein aggregates in models of Parkinson's disease, where inhibition of the autophagy-lysosome pathway leads to accumulation and potentiated toxicity of these aggregates in transgenic mice and neuronal cell cultures. Specifically, treatment with bafilomycin A1 enlarges proximity ligation assay-positive puncta associated with alpha-synuclein, confirming lysosomal involvement in its degradation and highlighting how enhancing this pathway could mitigate aggregate buildup. Low-dose bafilomycin (e.g., 10 nM) has shown protective effects by maintaining autophagic flux without complete blockade, attenuating neuronal death in models featuring autophagy-lysosome dysfunction akin to Parkinson's pathology. As of 2025, derivatives are under investigation to selectively modulate lysosomal proteolysis in organoid models of PD and AD. In models involving tau pathology, low-dose bafilomycin (10 nM) modulates to promote flux and support tau clearance without fully inhibiting the pathway, thereby reducing tau-induced vulnerability in neuronal cultures. This modulation preserves lysosomal function, counteracting the impaired autophagic turnover often observed in tauopathies, and underscores bafilomycin's utility in dissecting dose-dependent effects on protein degradation. In vivo studies demonstrate that intracerebral administration of bafilomycin A1 disrupts lysosomal acidification, leading to elevated amyloid-beta levels in brains, which indicates that intact activity normally facilitates amyloid-beta reduction through enhanced autophagic clearance. Recent investigations in organoids from 2023 to 2025 have utilized bafilomycin to model lysosomal failure in Parkinson's and Alzheimer's contexts, revealing impaired alpha- and handling in patient-derived and cerebral , with inhibition exacerbating collapse and aggregate propagation. These trials highlight bafilomycin's role in simulating neurodegenerative lysosomal deficits for therapeutic screening. From an immunosuppressant perspective, bafilomycin A1 reduces in neuroinflammatory settings by decreasing Iba1 expression and impairing cell survival under inflammatory stimuli in cultured , thereby attenuating pro-inflammatory responses linked to neurodegeneration. This effect stems from disrupted , which limits microglial phagocytic and inflammatory capacity without broadly suppressing neuronal autophagy.

Drug Interactions

Lysosomotropic Drug Interactions

Bafilomycin, a specific of the vacuolar H⁺-ATPase (), blocks proton translocation into the lysosomal lumen, resulting in elevated lysosomal pH and impaired lysosomal function. Lysosomotropic agents, such as , function as weak bases that diffuse across the lysosomal membrane and become protonated in the acidic environment, accumulating and buffering protons to similarly raise lysosomal pH. When combined, these agents produce additive effects on lysosomal alkalinization, exacerbating inhibition and leading to overload of the system, which promotes lysosomal membrane permeabilization (LMP) and release of hydrolytic enzymes into the . In experimental settings, co-administration of bafilomycin with or other weak bases significantly enhances accumulation compared to either agent alone, as indicated by increased LC3-II levels. These interactions are particularly valuable in research for validating inhibition through assays, where bafilomycin blocks activity and lysosomotropic agents like provide complementary neutralization to prevent autophagosome-lysosome fusion. By combining them, researchers can confirm a true accumulation of due to blocked degradation rather than upregulated synthesis, while minimizing off-target impacts such as altered endosomal trafficking from single-agent use at elevated concentrations. Standard protocols recommend short-term co-treatments (e.g., 2-4 hours) to assess accurately in mammalian cells.

Chemotherapeutic Synergies

Bafilomycin enhances the of chemotherapeutic agents by disrupting endosomal acidification, which reduces the of weakly basic drugs in acidic compartments and promotes their release into the and for greater therapeutic impact. In particular, it increases the intracellular retention of such as by inhibiting their trapping in endosomes and lysosomes, thereby elevating nuclear accumulation and in cancer cells. This mechanism has been demonstrated to potentiate doxorubicin's antiproliferative effects in hepatic carcinoma models. By overcoming multidrug resistance mechanisms involving lysosomal sequestration and efflux pumps, bafilomycin reverses resistance to chemotherapeutics trapped in lysosomal compartments. Treatment with bafilomycin alkalinizes lysosomes, reducing drug accumulation and restoring sensitivity to agents like in resistant cells, with combination effects showing strong synergy. V-ATPase inhibition, such as by bafilomycin, sensitizes cells to by enhancing . In , recent preclinical data from 2022 highlight synergistic growth inhibition when bafilomycin is combined with Src inhibitors like Si306, underscoring its potential in enhancing standard therapies such as through blockade.

Other In Vitro Interactions

Bafilomycin A1 shows synergistic activity with calcineurin inhibitors like FK506 against pathogenic fungi including , while FK506 synergizes with azoles like (FICI 0.25), potentially by disrupting vacuolar acidification and biosynthesis concurrently. Similarly, bafilomycin B1 synergizes with azoles like against glabrata, reducing minimum inhibitory concentrations by 4- to 16-fold through combined inhibition of and pathways, as demonstrated in assays. In cellular models of , bafilomycin A1 interacts antagonistically with the N-acetylcysteine () in H9c2 cardiomyocytes exposed to . While reduces formaldehyde-induced by mitigating (ROS), this protective effect is abolished by co-treatment with bafilomycin A1, which blocks autophagic flux and thereby prevents -mediated clearance of damaged organelles. Conversely, bafilomycin A1 exacerbates under these conditions, an effect attenuated by , highlighting autophagy's role as a downstream mediator of protection . Bafilomycin A1 also shows modulatory interactions with like rapamycin in various cell types, often used to dissect autophagic flux but revealing functional interplay. In induced pluripotent stem cells (iPSCs), rapamycin attenuates bafilomycin A1-induced by promoting upstream formation, countering the late-stage blockade and maintaining cellular , as evidenced by improved viability in co-treatment assays. In L6 myocytes, the combination enhances intramyocellular clearance via lipophagy, where rapamycin induces initiation while bafilomycin A1 prevents degradation, leading to accumulation that sensitizes cells to metabolic stress. These interactions underscore bafilomycin's utility in revealing pathway dependencies but also its potential to amplify or mitigate rapamycin's effects on cell survival and . Regarding apoptosis modulators, bafilomycin A1's cytotoxic effects are variably influenced by caspase inhibitors like Z-DEVD-FMK across cell lines. In pediatric B-cell cells, bafilomycin A1 triggers that is not rescued by Z-VAD-fmk, indicating a caspase-independent mechanism involving lysosomal dysfunction. However, in H9c2 cardiomyocytes under formaldehyde stress, Z-DEVD-FMK blocks bafilomycin A1-enhanced , suggesting context-dependent crosstalk between autophagy inhibition and caspase activation .

References

  1. [1]
    Structural diversity and biological activities of naturally derived ...
    Apr 8, 2025 · Bafilomycins (Baf) are plecomacrolide antibiotics characterized by a 16-membered macrocyclic lactone containing two sets of conjugated dienes ...
  2. [2]
    Chemistry and structure activity relationships of bafilomycin A1, a ...
    Bafilomycin A1, a macrolide antibiotic isolated from the fermentation of Streptomyces spp., is a potent and selective inhibitor of vacuolar-type proton ...
  3. [3]
  4. [4]
    Bafilomycin A1 Inhibits HIV-1 Infection by Disrupting Lysosomal ...
    Aug 29, 2024 · Our data suggest that bafilomycin A1 inhibits HIV-1 replication in part by disrupting the lysosomal cholesterol trafficking pathway.
  5. [5]
    Genome-directed discovery of antiproliferative bafilomycins from a ...
    Since the first discovery of bafilomycin biosynthetic gene cluster (BGC) from Streptomyces hygroscopicus ATCC31955 in 2012 [1], multiple homologous BGCs ...Missing: Bafilomycin | Show results with:Bafilomycin
  6. [6]
  7. [7]
    Characterization of the Bafilomycin Biosynthetic Gene Cluster ... - NIH
    This compound was first isolated from Streptomyces griseus sp. sulphurus (TÜ 1922) in 1984. ... The term plecomacrolide was later introduced to describe a ...Missing: 1983 | Show results with:1983
  8. [8]
    Characterization of bafilomycin biosynthesis in Kitasatospora setae ...
    Mar 15, 2017 · An analysis of gene clusters from K. setae KM-6054 governing the biosynthesis of bafilomycins revealed that it contains five large open reading frames (ORFs)
  9. [9]
  10. [10]
    Engineering Bafilomycin High-Producers by Manipulating ... - NIH
    Jan 11, 2021 · Since bafilomycin A1, B1, and C1 were first isolated from Streptomyces griseus sp. sulphurus (TÜ 1922) in 1983 [5], nearly thirty ...Missing: paper | Show results with:paper
  11. [11]
    Metabolic products of microorganisms. 224. Bafilomycins ... - PubMed
    The bafilomycins A1, A2, B1, B2, C1 and C2, a new type of macrolide antibiotics with a 16-membered lactone ring, were isolated from the fermentation broth ...
  12. [12]
    Bafilomycin A1 | C35H58O9 | CID 6436223 - PubChem
    Bafilomycin A1 is the most used of the bafilomycins, a family of toxic macrolide antibiotics derived from Streptomyces griseus.
  13. [13]
    A conformational study of bafilomycin A1 by x-ray crystallography ...
    We report the structure determination of bafilomycin A1 by X-ray crystallography and a reassignment of the 1H and 13C NMR spectra. By the measurement of 13C ...
  14. [14]
    Bafilomycin A1: Uses, Interactions, Mechanism of Action - DrugBank
    Aug 25, 2010 · Structure for Bafilomycin A1 (DB06733). ×. Close. Weight: Average: 622.84. Monoisotopic: 622.408083448; Chemical Formula: C35H58O9. Synonyms.
  15. [15]
    [PDF] PRODUCT INFORMATION - Cayman Chemical
    Bafilomycin A1 is soluble in organic solvents such as DMSO and methanol. The solubility of bafilomycin A1 in these solvents is approximately 5 mg/ml ...Missing: logP | Show results with:logP
  16. [16]
    BAFILOMYCIN A1 CAS#: 88899-55-2 - ChemicalBook
    Water Solubility, Soluble in methanol, ethanol, acetone and chloroform. Insoluble in water. ; BRN, 4730700 ; Stability, Stable for 2 years from date of purchase ...
  17. [17]
  18. [18]
    Bafilomycin A1 | CAS 88899-55-2 | P212121 Store
    It is soluble in ethanol, methanol and DMSO, and is light-sensitive, especially while in solution. It should be stored at below 20 degrees C. The ...
  19. [19]
    Bafilomycin A1 - Endosomal Acidification Inhibitor | InvivoGen
    Bafilomycin A1 (BafA1), a macrolide antibiotic isolated from Streptomyces species, is a specific vacuolar H + ATPase (V-ATPase) inhibitor.Missing: discovery griseus 1983 Kyowa Hakko Kogyo
  20. [20]
    Total Synthesis of Bafilomycin A1 - Kleinbeck - Chemistry Europe
    Feb 16, 2012 · A convergent synthesis of bafilomycin A1, a potent inhibitor of V-type ATPases, is presented. The synthesis relies on the zinc triflate ...
  21. [21]
  22. [22]
    Synthesis and structure-activity relationships of bafilomycin A1 ...
    May 21, 1998 · The macrolide antibiotic bafilomycin A1 is a highly potent and selective inhibitor of all the vacuolar ATPases (V-ATPases).Missing: elucidation paper
  23. [23]
    inhibition by bafilomycins A1, B1, C1 and D - PubMed
    Bafilomycin A1 displays the highest activity, followed by bafilomycin B1, C1 and D. The different potency of bafilomycins correlates with their ability to inh …Missing: family | Show results with:family
  24. [24]
    Bioactive Bafilomycins and a New N-Arylpyrazinone Derivative from ...
    Bioactive Bafilomycins and a New N-Arylpyrazinone Derivative from Marine-derived Streptomyces sp. ... bafilomycin D, 9-hydroxybafilomycin D, and bafilomycin A1.
  25. [25]
  26. [26]
    Vacuolar ATPases: rotary proton pumps in physiology and ... - Nature
    The vacuolar (V-)ATPases are ATP-driven proton pumps that function to acidify intracellular compartments and transport protons across the plasma membrane.Missing: structure | Show results with:structure
  27. [27]
  28. [28]
    Molecular basis of V-ATPase inhibition by bafilomycin A1 - PMC
    Mar 19, 2021 · The structure reveals six bafilomycin A1 molecules bound to the c-ring. One bafilomycin A1 molecule engages with two c subunits and disrupts the ...
  29. [29]
    Inhibitors of V-ATPases: old and new players
    Feb 1, 2009 · The discovery of the plecomacrolide bafilomycin as the first specific and highly potent inhibitor of V-ATPases(Bowman et al., 1988) opened a new ...
  30. [30]
    Bafilomycin A1 | H+-ATPase - Tocris Bioscience
    Rating 4.3 (3) · $45 deliverySolubility Data for Bafilomycin A1. Solvent, Max Conc. mg/mL, Max Conc. mM. Solubility. DMSO, 4.98, 8. Preparing Stock Solutions for Bafilomycin A1. The ...Missing: water | Show results with:water
  31. [31]
    Cryo-EM structures of intact V-ATPase from bovine brain - Nature
    Aug 6, 2020 · We report two cryo-EM structures of the intact V-ATPase from bovine brain with all the subunits including the subunit H, which is essential for ATPase activity.
  32. [32]
    Guidelines for the use and interpretation of assays for monitoring ...
    Feb 8, 2021 · Here, we present a set of guidelines for investigators to select and interpret methods to examine autophagy and related processes.
  33. [33]
    The Autophagy Inhibitor Bafilomycin Inhibits Antibody-Dependent ...
    Jun 28, 2025 · These findings show that autophagy inhibition by bafilomycin confers ADCC resistance by altering apoptosis, immune signaling, and HER2 dynamics.Missing: 2024 | Show results with:2024
  34. [34]
    Bafilomycin-A1 and ML9 Exert Different Lysosomal Actions to Induce ...
    Furthermore, both the drugs target lysosomes through different mechanisms. For the latter, cell death is likely due to lysosomal membrane permeabilization and ...<|separator|>
  35. [35]
    Cathepsin D Triggers Bax Activation, Resulting in Selective ...
    We demonstrated that Cat D, an aspartic protease released from the lysosomes into the cytosol, is an upstream trigger of Bax activation.
  36. [36]
    Lysosomal membrane permeabilization and cathepsin release is a ...
    Lysosomal membrane permeabilization and cathepsin release is a Bax/Bak-dependent, amplifying event of apoptosis in fibroblasts and monocytes. Cell Death ...Missing: bafilomycin | Show results with:bafilomycin
  37. [37]
    Bafilomycin A1 induces caspase-independent cell death in ... - Nature
    Nov 15, 2016 · Bafilomycin A1 induces apoptosis in PC12 cells independently of intracellular pH. ... apoptosis—modulatory effect of p38 MAPK and JNK inhibitors.
  38. [38]
    [PDF] Bafilomycin A1 induces caspase-dependent apoptosis and in
    Jul 22, 2021 · Meanwhile, two classical apoptosis markers, caspase-3 activation and Poly (ADP-ribose) polymerase (PARP) cleavage, were observed in bafilo-.
  39. [39]
    Inhibition of macroautophagy by bafilomycin A1 lowers proliferation ...
    Aug 6, 2025 · Inhibition of autophagy by bafilomycin-A1 also phosphorylates ERK-JNK-p38, thereby facilitates the cell cycle arrest and caspase-dependent ...<|separator|>
  40. [40]
    Bafilomycin A1 inhibits the growth and metastatic potential of ... - NIH
    The V-ATPase is typically highly activated in cancer cells, in which it has the following functions: Regulating cell proliferation, apoptosis and autophagy; ...Missing: 50-200 | Show results with:50-200
  41. [41]
    Bafilomycin A1 induces colon cancer cell death through impairment ...
    Feb 12, 2025 · The results showed that CRC cells were much more sensitive to low concentrations of BA1 than lung cancer cells and normal colon fibroblast cells ...
  42. [42]
  43. [43]
    TMEM175 Is an Organelle K+ Channel Regulating Lysosomal ...
    Aug 27, 2015 · TMEM175 regulates lysosomal membrane potential and pH stability. Knocking out TMEM175 leads to abnormal autophagosome-lysosome fusion.Article · Results · Tmem175 Is A Novel Protein...
  44. [44]
    Effects of bafilomycin A1, a vacuolar type H+ ATPase inhibitor, on ...
    Methods: This study investigated the effects the bafilomycine A1, an inhibitor of the vacuolar type H+ATPase and the EIPA, an inhibitor of the Na+/H+ exchanger ...Missing: compensatory activation
  45. [45]
    Regulation of Vacuolar pH and Its Modulation by Some Microbial ...
    Na+ accumulated in the lumen by the Na+/K+-ATPases could in turn drive Na+/H+ exchange, promoting acidification. (B) Schematic representation of the pHs of the ...
  46. [46]
  47. [47]
    Lysosome Inhibitors Enhance the Chemotherapeutic Activity of ...
    Our findings suggest that bafilomycin A1 and chloroquine potentiate the anticancer effect of doxorubicin in hepatic cancer cells.Missing: synergy | Show results with:synergy
  48. [48]
    Autophagy Inhibition Enhances Anti-Glioblastoma Effects of ... - MDPI
    Combined treatments of Si306 and pro-Si306 with bafilomycin A1 were synergistic in nature, and the inhibition of autophagy sensitized glioblastoma cells to Src ...Autophagy Inhibition... · 3. Results · 3.4. Bafilomycin A1 Enhances...
  49. [49]
    Inhibiting autophagy increases epirubicin's cytotoxicity in breast ...
    Epirubicin treatment of both cell lines revealed an IC50 at 48 h of 3.0 μM in MDA‐MB‐231 cells and 2.0 μM in SK‐BR‐3 cells. Bafilomycin A1 (5, 10, and 20 nM) ...Propidium Iodide Staining... · Caspase‐3 And Caspase‐9... · Bafilomycin A1 Enhanced...Missing: IC50 | Show results with:IC50<|separator|>
  50. [50]
    The impact of tumour pH on cancer progression: strategies for ...
    The acidic microenvironment within a tumour can promote migration, invasion and metastasis of cancer cells through a variety of mechanisms.
  51. [51]
    Alpha-synuclein aggregation involves a bafilomycin A1-sensitive ...
    ALP inhibition using bafilomycin A 1 (BafA1) significantly potentiates toxicity of aggregated α-synuclein species in transgenic mice and in cell culture.
  52. [52]
    synuclein aggregates in Parkinson's disease using the AUTOTAC ...
    Jun 24, 2023 · These PLA+ puncta were further enlarged upon autophagic inhibition using bafilomycin A1, indicative of lysosomal degradation (Fig. 3L and N). As ...
  53. [53]
    Low-dose bafilomycin attenuates neuronal cell death ... - PubMed
    Jun 1, 2010 · Our findings suggest that low-dose bafilomycin is cytoprotective in part through its maintenance of the autophagy-lysosome pathway.
  54. [54]
    Prolonged tau clearance and stress vulnerability rescue by ... - Nature
    Jun 26, 2020 · In these models, autophagy activators reduce the levels of misfolded and aggregated proteins, mitigating the spreading of tau and neuronal loss ...
  55. [55]
    Tau deletion impairs intracellular β-amyloid-42 clearance and leads ...
    Nov 10, 2014 · Bafilomycin-A1 significantly increased Aβ1-42 (n = 5, p < 0.045) compared to DMSO. In the presence of Bafilomycin-A1, Nilotinib was unable to ...
  56. [56]
    Disruption of lysosomal proteolysis in astrocytes facilitates midbrain ...
    Jan 10, 2024 · Disruption of lysosomal proteolysis in astrocytes facilitates midbrain organoid proteostasis failure in an early-onset Parkinson's disease model.
  57. [57]
    Neuronal Autophagy Failure Drives α-Synuclein Transfer to ...
    Jun 14, 2025 · Impaired autophagy is a molecular hallmark of neurodegenerative diseases (Wilson et al, 2023). The clearance of aggregate-prone proteins, such ...
  58. [58]
    Bafilomycin 1A Affects p62/SQSTM1 Autophagy Marker Protein ...
    Jul 29, 2024 · Autophagy can also intricately influence inflammation and microglial activation, suggesting that promoting the early stages of autophagy ...
  59. [59]
    Lysosomal acidification dysfunction in microglia: an emerging ...
    Aug 5, 2023 · Defective microglial lysosomal acidification leads to impaired phagocytic and autophagic functions which result in the perpetuation of neuroinflammation.
  60. [60]
    Methods in Mammalian Autophagy Research - ScienceDirect.com
    Feb 5, 2010 · Final degradation of autophagic cargo inside autolysosomes can also be inhibited by ammonium chloride, bafilomycin A1, and lysosomal protease ...
  61. [61]
    Lysosomal agents inhibit store-operated Ca 2+ entry
    Jan 27, 2021 · Probing lysosomes in another manner, we specifically inhibited the lysosomal H+ pump (V-ATPase) with bafilomycin A1, to increase the lysosomal ...
  62. [62]
    Acetylated microtubules are required for fusion of autophagosomes ...
    Nov 22, 2010 · The treatments resulted in a 3-fold increase of LC3I levels ... bafilomycin A1 and ammonium chloride (NH4Cl) were purchased from Sigma.<|control11|><|separator|>
  63. [63]
    Dynamics of the Degradation of Ubiquitinated Proteins by ...
    Autophagy inhibitors bafilomycin A1, ammonium chloride, and 3-methyladenine failed to increase ubiquitinated protein levels. The proteasome inhibitor epoxomicin ...<|control11|><|separator|>
  64. [64]
    Untangling Autophagy Measurements | Circulation Research
    Jan 30, 2015 · Several agents are suitable to achieve lysosomal blockade: bafilomycin A1, chloroquine, and ammonium chloride raise the intralysosomal pH, ...
  65. [65]
    Pharmacological CDK4/6 inhibition promotes vulnerability to ...
    Feb 10, 2025 · Altogether, these data suggest that sequential treatment with abemaciclib and lysosomotropic agents selectively achieves toxicity in HR+ breast ...
  66. [66]
    Lysosome Inhibitors Enhance the Chemotherapeutic Activity of ...
    Oct 21, 2016 · The plots show synergistic cell killing between doxorubicin and bafilomycin A1/chloroquine (combination) in L02 and HepG2 cell lines (fig. 2 ...
  67. [67]
    Inhibition of endosomal sequestration of basic anticancer drugs - NIH
    Chloroquine, omeprazole and bafilomycin A showed concentration-dependent effects to raise endosomal pH, and to inhibit sequestration of doxorubicin in endosomes ...
  68. [68]
    Influence of lysosomal sequestration on multidrug resistance ... - NIH
    A possible mechanism to reverse lysosomal drug accumulation of weak chemotherapeutic bases is by treatment with lysosome alkalinizing agents such as bafilomycin ...
  69. [69]
    Selective inhibition of tumor cell associated Vacuolar-ATPase 'a2 ...
    Interestingly, bafilomycin treated cisplatin ... The cisplatin sensitization was more pronounced in the cisplatin resistant ovarian cancer cells than cisplatin ...
  70. [70]
    Si306 and pro-Si306 in combination with bafilomycin A1 (Baf A1 ...
    Si306 and pro-Si306 in combination with bafilomycin A1 (Baf A1) have a synergistic effect on growth inhibition of glioblastoma cells. (a) The sensitivity of ...
  71. [71]
    Synergistic Antifungal Activities of Bafilomycin A1, Fluconazole, and ...
    We demonstrate that FK506 exhibits marked synergistic activity with the H+ATPase inhibitor bafilomycin A1 via a novel action distinct from calcineurin loss of ...Missing: paper | Show results with:paper
  72. [72]
    Vacuolar proton-translocating ATPase is required for antifungal ...
    Jan 23, 2019 · In the current study, we demonstrated that the plecomacrolide V-ATPase inhibitor bafilomycin B1 exerts a synergistic effect with azole ...Missing: vitro | Show results with:vitro
  73. [73]
    Interactions between oxidative stress, autophagy and apoptosis in ...
    Inhibitors of oxidative stress (N-acetylcysteine, NAC), autophagy (bafilomycin A1, Baf) and apoptosis (Z-DEVD-FMK) were used to investigate their interactions.
  74. [74]
    Reversal of intramyocellular lipid accumulation by lipophagy and a ...
    Aug 22, 2016 · Combination of rapamycin and bafilomycin treatment suggest a new mechanism for clearing intramyocellular lipids. L6 myocytes were treated with ...
  75. [75]
    Bafilomycin A1 targets both autophagy and apoptosis pathways in ...
    Mar 1, 2015 · We show that bafilomycin A1 at a low concentration (1 nM) effectively and specifically inhibited and killed pediatric B-cell acute lymphoblastic leukemia cells.Missing: structural B1 C1