WO2020154359A1 - Cofilin phosphorylation for cancer treatment - Google Patents

Cofilin phosphorylation for cancer treatment Download PDF

Info

Publication number
WO2020154359A1
WO2020154359A1 PCT/US2020/014542 US2020014542W WO2020154359A1 WO 2020154359 A1 WO2020154359 A1 WO 2020154359A1 US 2020014542 W US2020014542 W US 2020014542W WO 2020154359 A1 WO2020154359 A1 WO 2020154359A1
Authority
WO
WIPO (PCT)
Prior art keywords
cofilin
cells
patient
cell
hiv
Prior art date
Application number
PCT/US2020/014542
Other languages
French (fr)
Inventor
Yuntao Wu
Hong SHANG
Yajing FU
Original Assignee
Virongy L.L.C.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Virongy L.L.C. filed Critical Virongy L.L.C.
Publication of WO2020154359A1 publication Critical patent/WO2020154359A1/en
Priority to US18/118,409 priority Critical patent/US20230248907A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56983Viruses
    • G01N33/56988HIV or HTLV
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • ART Antiretroviral therapy
  • the natural course of HIV infection leads to multiple CD4 T cell defects (1), including the impairment of T cell migration and homing to lymphoid tissues such as GALT (Gut-Associated Lymphoid Tissues) (2-4).
  • GALT Guard-Associated Lymphoid Tissues
  • T cells such as CD8 T cells play a critical role in cancer immunity, but tumors can use various defense mechanisms, either directly or indirectly, to inhibit T migration to tumor bed for immune control (11).
  • T cell activity is mainly regulated by receptor signaling.
  • Persistent stimulation and signaling through chemotactic receptors e.g., chemokine receptors, cytokine receptors, integrin receptors, adhesion molecule receptors, and co-stimulatory molecule receptors such as CD28, CTLA-4, and PD-l/PD-L
  • chemotactic receptors e.g., chemokine receptors, cytokine receptors, integrin receptors, adhesion molecule receptors, and co-stimulatory molecule receptors
  • CD28 e.g., CD28, CTLA-4, and PD-l/PD-L
  • co-stimulatory molecule receptors such as CD28, CTLA-4, and PD-l/PD-L
  • chemokine receptors e.g., chemokine receptors, cytokine receptors, integrin receptors, adhesion molecule receptors, and co-stimulatory molecule receptors
  • HIV signaling through CXCR4 has been shown to activate an actin depolymerizing factor, cofilin, to promote the actin dynamics necessary for viral nuclear entry in blood resting CD4 T cells (9, 16).
  • an actin depolymerizing factor cofilin
  • cofilin actin depolymerizing factor
  • the persistent stimulation of immune cells by high levels of inflammatory chemokines, such as IP-10 (17, 18), and tumor antigens can also cause T polarization and dysfunction, and impair T cell motility (Fig. 1)
  • Cofilin is an actin-binding protein, one of the membranes of the ADF/cofilin family proteins ubiquitously present among eukaryotes. ADF/cofilin proteins bind and depolymerize filamentous F-actin in a pH-dependent manner and are responsible for the high turnover rates of actin filaments in cells. The first member of the ADF/cofilin family proteins was identified in extracts of embryonic chick brain (19). Cofilin was later purified from porcine brain and named cofilamentous structures with actin, or cofilin for short.
  • ADF actin depolymerizing factor
  • destrin actin depolymerizing factor
  • cofilin 1 non-muscle cofilin or cofilin
  • cofilin-2 muscle-cofilin
  • human cofilin possesses a core consisting of a five-stranded mixed b- sheet (b ⁇ to b5).
  • the first four strands (b ⁇ to b4) are anti-parallel, and b5 runs parallel to b4 and anti -parallel to b3.
  • the residues 159-161 at the C-terminal form a short stand, b6.
  • Five helices (al to a.5) surround the central b-sheet, and a salt bridge is formed between His 133 and Asp 98 that may influence the pH sensitivity of cofilin in actin binding and depolymerization.
  • Cofilin binds to G actin, and the binding site is located in a region centered around Tyr 117 , which includes several residues in a.4 (such as Lys 112 , Lys 114 , Met 115 , and He 124 ) and b5. In addition, a few other residues such as He 12 , Pro 110 and Leu 128 are also suggested in G-actin binding. Cofilin also binds to F-actin, and this requires two sites on cofilin, a G site that interacts with actin subdomains 1 and 3 and an F site that interacts with actin subdomains 1 and 2.
  • Residues important for F-actin binding on cofilin have been identified through mutagenesis and include several residues in b4 (such as Lys 96 and Asp 98 ) and a.5 (such as Glu 151 , Lys 152 , and Gly 155 ). Other regions including b3 and b5 may additionally be involved in F-actin binding (21).
  • Cofilin depolymerizes actin filaments through two mechanisms: direct severing and increasing the off-rate of actin subunits from the (-) end (22, 23).
  • Cofilin has a higher affinity for ADP-actin than for ATP-actin, and cofilin binding to actin filaments facilitates phosphate dissociation from ATP-actin.
  • Hydrolysis of ATP increases cofilin binding affinity, changing the twist of the actin helix and severing actin filaments to shorter segments (24).
  • These severed actin filaments can increase the speed of actin polymerization by providing more free (+) ends for nucleation by the Arp2/3 complex (25).
  • cofilin and the Arp2/3 complex work together to regulate actin treadmilling, in which ATP-actin is preferentially incorporated into the filaments through Arp2/3 at the (+) end, and then hydrolyzed into “older” ADP-actin and dissociated from the (-) end by cofilin (23, 26).
  • cofilin serine 3 In cells, the activity of cofilin is mainly regulated by phosphorylation of serine 3 at the N-terminal, which inhibits cofilin binding to G-actin and F-actin.
  • the kinases responsible for cofilin serine 3 phosphorylation are the LIM domain kinases (LIMK) and Tes kinases, which are targets of the Rho family GTPases such as Rho, Rac, and Cdc42 (27).
  • Rho family GTPases activate PAK1, PAK2, PAK4, ROCK, or MRCKa (myotonic dystrophy kinase- related Cdc42-binding protein kinase) which then activate LIMK through direct
  • Cofilin is activated through dephosphorylation of serine 3 by phosphatases such as PP1, PP2A, slingshot IL (SSH1L), and chronophin, which couple cofilin activity to different signal pathways (28-30).
  • phosphatases such as PP1, PP2A, slingshot IL (SSH1L), and chronophin, which couple cofilin activity to different signal pathways (28-30).
  • SSH1L slingshot IL
  • Y68 tyrosine 68
  • cycles of cofilin phosphorylation and dephosphorylation are required to sustain the actin dynamics essential for driving directional cell migration.
  • cofilin is a key regulator of actin dynamics and is also involved in multiple cellular processes (32). The ability of cofilin to modulate actin polymerization or
  • cofilin has also been shown to mediate actin nuclear localization, which may be involved in the regulation of gene expression.
  • Actin and actin-related proteins such as Arp7, Arp9, and Baf53 are parts of the chromatin-remodeling complex RSC and SWPSNF (35, 36). Actin is also part of the pre initiation complexes and is necessary for transcription by RNA polymerase II (37).
  • Cofilin- mediated actin nuclear localization may serve to connect the cytoskeletal processes to chromatin remodeling and the initiation of transcription.
  • PLD1 phospholipase D1
  • Cofilin directly and specifically interacts with PLD1 upon phosphorylation by LIMK1.
  • Phospho- cofilin also stimulates PLD1 activity, suggesting that phospho-cofilin may control a variety of cellular functions by its stimulatory effect on PLD1 (38).
  • cofilin plays important roles in regulating T cell migration, chemotaxis, and T cell activation.
  • cofilin is required for the formation of the supramolecular activation clusters critical for sustaining signaling and T cell activation.
  • Cell-permeable peptides that block cofilin interaction with F- actin impair receptor capping and immunological synapse formation, resulting in inhibition of T cell activation (39).
  • cofilin exists largely as the serine 3 phosphorylated form. T cell activation or chemotactic stimulation leads to transient cofilin activation by
  • CD2 costimulatory receptors
  • CD28 CD28
  • chemokine receptors such as CXCR4.
  • TCR/CD3 stimulation activates the Arp2/3 complex for actin polymerization
  • CD28- mediated costimulation triggers cofilin activation, which is required for dynamic actin reorganization and sustaining T cell signaling (40).
  • cortical actin in resting T cells is relatively static in the absence of T cell activation or chemotactic stimulation. This lack of actin activity limits viral early processes such as entry, DNA synthesis, and nuclear migration (9).
  • Cofilin increases cortical actin dynamics and actin treadmilling, facilitating viral intracellular migration towards the nucleus (9).
  • Cofilin and actin-mediated HIV nuclear localization is suggested to be essential for the establishment of HIV-1 latency in resting CD4 T cells (9, 16). Slight inhibition of cofilin expression through siRNA knockdown increases cortical actin density, which leads to an increase in HIV DNA synthesis but a decrease in the amounts of HIV- 1 nuclear DNA and early transcripts (9).
  • HIV-l-mediated cofilin activation in resting CD4 T cells is shown to be through the God-dependent signaling from CXCR4; pertussis toxin (PTX), a bacterial toxin inhibiting G- protein-coupled receptors by the ADP-ribosylation of God, inhibits cofilin activation and HIV-1 latent infection of resting T cells (9).
  • Cofilin can be phosphorylated by LIMK, and HIV-1 binding to blood CD4 T cells and macrophages triggers rapid activation of LIMK1, coincident with HIV-mediated early actin polymerization in T cells (42).
  • LIMKl/cofilin activity to directly regulate actin and CXCR4 dynamics critical for viral entry, postentry DNA synthesis, and nuclear migration.
  • Inhibition of LIMK1 activity through siRNA knockdown decreases filamentous actin and T cell chemotaxis towards SDF- 1.
  • the decrease in cortical actin density also leads to an increase in CXCR4 internalization and surface recycling.
  • LIMK-mediated early cortical actin polymerization may result in a temporary block to CXCR4 internalization, facilitating viral fusion and CXCR4 signaling.
  • the LIMK1 knockdown cells also support lower viral entry, DNA synthesis, and nuclear migration.
  • transient treatment of resting CD4 T cells with a pharmacological agent, okadaic acid activates LIMK and promotes HIV latent infection of resting CD4 T cells.
  • the signaling pathway that mediates LIMK activation by HIV-1 is identified as the Racl, PAK1/2 and LIMK pathway in blood resting T cells. The activation is likely triggered by gpl20 signaling through both CD4 and CXCR4, as well as from both Gori and Gaq (42).
  • HIV binding to CD4 and CXCR4 is also suggested to trigger filamin A activation which then leads to RhoA-ROCK-LIMK activation.
  • This activity is proposed to permit F-actin reorganization for receptor clustering (45).
  • HIV gpl20-mediated cofilin phosphorylation is also suggested to inhibit T cell chemotaxis towards SDF-1 (46).
  • Nef another HIV pathogenic factor, Nef, is also suggested to regulate cofilin activity (47).
  • Over-expression of Nef in a human cancer Jurkat T cell inhibits SDF-1 -induced membrane ruffling, actin rearrangement, and cell migration towards
  • Nef-mediated cofilin dysregulation may affect migratory behavior of infected T cells (47).
  • compositions, kits, and the like for determining the levels of cofilin phosphorylation in blood T cells (CD4 or CD8), which can be used to quantify the immune damages caused by chronic viral infection and cancer, and to predict the recovery of T cells following treatment. Based on this determination of a correlation between cofilin phosphorylation and immune dysfunction, suitable patients may be treated with a composition that causes cofilin dysregulation and restores cofilin phosphorylation.
  • levels of cofilin phosphorylation in HIV-infected patients can be used as a marker to quantify immune dysfunction and damages by the virus.
  • levels of cofilin phosphorylation in HIV-infected patients can be used to measure drug effects on immune function (ART and other immune modulating drugs).
  • levels of cofilin phosphorylation in HIV-infected patients can be used to predict the recovery of T cell functions from drug treatment (ART and other immune modulating drugs).
  • levels of coflin phosphorylation in cancer cells can be used to quantify alteration in cell motility, migration, tissue infiltration, and metastasis.
  • levels of cofilin phosphorylation in T cells (CD4/CD8) in cancer patients can be used to predict immune dysfunction in cancer, and predict drugs effects in restoring anti-tumor immunity.
  • a method for detecting and treating immune dysfunction in a patient comprises (a) quantifying cofilin phosphorylation levels in T cells (CD4/CD8), monocytes/macrophages, B cells, natural killer (NK) cells, and/or cancer cells of said patient, wherein said patient with immune dysfunction has lower or higher cofilin phosphorylation levels than a control healthy person or patient; and (b) administering to said patient an effective amount of a composition that corrects cofilin dysregulation and restores cofilin phosphorylation.
  • the patient has a chronic disease causing immune dysfunction.
  • the said chronic disease is HIV infection or cancer.
  • said immune dysfunction is aberrant T cell, macrophage, B cell, NK cell activation, impairment of T cell, monocyte/macrophage, B cell, NK cell motility and/or tissue migration.
  • the composition is a cofilin phosphatase inhibitor or cofilin kinase activator. In some embodiments, the composition comprises okadaic acid.
  • a method for determining immune dysfunction in a patient comprises quantifying cofilin phosphorylation in T cells (CD4/CD8),
  • monocyte/macrophages B cells, natural killer cells, and/or cancer cells in said patient.
  • the patient is a cancer patient or HIV-infected patient.
  • the method for treating cancer in a patient comprises (a) quantifying cofilin phosphorylation levels in T cells (CD4/CD8), monocyte/macrophages, B cells, natural killer cells, and/or cancer cells of said patient, wherein said patient has lower or higher cofilin phosphorylation levels than a control healthy person; and (b) administering to said patient an effective amount of a composition that restores cofilin phosphorylation.
  • the composition is a cofilin phosphatase inhibitor or cofilin kinase activator or an antibody against chemotactic receptors that can trigger cofilin phosphorylation or dephosphorylation or a small molecule that can trigger cofilin
  • the composition comprises okadaic acid.
  • the method is for treating a patient having cancer, including, for example, patients who do not have HIV or AIDS.
  • FIGURE 1 Persistent stimulation of T cell surface receptors can lead to T cell polarization to different lineages.
  • A Persistent and chronic stimulation of chemotactic receptors (e.g., chemokine receptors, cytokine receptors, integrin receptors, adhesion molecule receptors, and co-stimulatory molecule receptors, such as CD28/B7, B7-2, CTLA- 4/B7, B7-2, and PD-l/PD-L) can leads to T cell polarization and commitment to distinct lineages, such as an irreversible pathogenic lineage with chronic cofilin hyperactivation.
  • B The signaling pathways involved in cofilin activation through the stimulation of receptors.
  • FIGURE 2 Cofilin hyperactivation in HIV infection.
  • A Flowchart of the clinical study.
  • B Development of the reverse-phase cofilin microarray for profiling cofilin phosphorylation. Synthetic peptides or cell lysates were serially diluted (1 : 1) and printed onto the microarray slides, which were then stained with antibodies against either total cofilin (right) or phospho-cofilin (left). P-cofilin-S3, a synthetic cofilin peptide with serine 3 phosphorylated; cofilin-S3, a similar peptide with no serine 3 phosphorylation.
  • A431 or HeLa cells were not treated or treated with human epithelial growth factor (EGF) or pervanadate (Perv).
  • C Relative levels of p-cofilin in blood resting CD4 T cells from HIV-infected patients with ART (HIV+ART) or without ART (HIV), or healthy control donors (HC) were profiled. Box plots show interquartile range, median, and range. There were no statistically significant differences in the total protein levels of the resting CD4 T cells from HC, HIV, and HIV+ART (see Materials and Methods).
  • D and E The correlation between levels of p- cofilin and plasma viral load (D) and CD4 T cell count (E) in untreated patients were plotted using Spearman rank correlation tests (Ln, natural logarithm).
  • FIGURE 3 Quantification of effects of cofilin hyperactivation on T cell migration.
  • A A3R5.7 T cells were treated with different dosages of R10015 for one hour. Phospho-cofilin and total cofilin were quantified by Western blot.
  • C R10015 inhibits cofilin phosphorylation and T cell chemotaxis in response to CXCL12.
  • A3R5.7 cells were treated with different dosages of R10015 for one hour, and then added to the upper chamber of a 24-well transwell plate.
  • D The linear correlation between T cell migration and levels of cofilin phosphorylation.
  • the X-axis is the relative ratio of p-cofilin/cofilin derived from (B); the Y- axis is the number of migrating cells derived from (C).
  • FIGURE 4 Cofilin hyperactivation in blood cancer cells.
  • Peripheral blood mononuclear cells PBMC from healthy donors, Jurkat cancer T lymphocyte cells (derived from a patient with acute T cell leukemia), A3R5.7 T cells (derived from the peripheral blood huffy coat of a four-year-old patient with acute lymphoblastic leukemia), or THP-1 blood monocytic cancer cells (derived from an acute monocytic leukemia patient) were fixed, permeabilized, washed, and then stained with a rabbit polyclonal anti-human p-cofilin antibody for 60 min. Cells were washed twice and stained with Alexa Fluor 488-labeled anti rabbit antibodies.
  • PBMC Peripheral blood mononuclear cells
  • Jurkat cancer T lymphocyte cells derived from a patient with acute T cell leukemia
  • A3R5.7 T cells derived from the peripheral blood huffy coat of a four-year-old patient with acute lymphoblastic leukemia
  • FIGURE 5 Cofilin dysregulation in blood CD8 T cells and monocytes of cancer patients.
  • Peripheral blood cytotoxic T cells (CD8 T cells) or monocytes from health donors or colorectal cancer patients were fixed, permeabilized, washed, and then stained with a rabbit polyclonal anti-human p-cofilin antibody for 60 min. Cells were washed twice and stained with Alexa Fluor 488-labeled anti-rabbit antibodies. Cells were washed twice, and then analyzed by flow cytometry. The relative p-cofilin staining was calculated. (A and B) shows the representative p-cofilin staining from 8 donors.
  • the filled grey plots are isotype staining, the red line plots are p-cofilin staining.
  • C The relative P-cofilin staining was calculated by using the mean fluorescent intensity from p-cofilin staining.
  • P-cofilin was similarly analyzed in blood monocytes of cancer patients and healthy donors.
  • FIGURE 6 Characteristics of clinical study participants. Details are in examples.
  • FIGURE 7 Patient enrollment and grouping. Details are in examples.
  • a major immune dysfunction persisting in chronic diseases such as HIV infection and cancer is the impairment of T cell motility and migration to tissues.
  • Therapeutics such as antiretroviral therapy or cancer therapy frequently do not fully restore T cell motility for tissue migration and infiltration.
  • Cofilin is an actin-depolymerizing factor that regulates actin dynamics for T cell migration.
  • the present inventors determined that the levels of cofilin phosphorylation in blood T cells (CD4 or CD8) can be used to quantify the immune damages caused by chronic viral infection and cancer, and to predict the recovery of T cells following treatment. Based on this determination of a correlation between cofilin phosphorylation and immune dysfunction, suitable patients may be treated with a composition that corrects cofilin dysregulation and restores cofilin phosphorylation.
  • cells can be treated with cofilin phosphatase inhibitors or cofilin kinase activator to increase cofilin phosphorylation, as demonstrated by the use of okadaic acid to increase cofilin phosphorylation (42) or an anti- integrin antibody to modulate the cofilin pathway (49).
  • a potential pathogenic role of HIV- 1 -mediated cofilin dysregulation is proposed from studies of cofilin activation in blood resting CD4 T cells treated with HIV or gpl20 (50). It was shown that HIV-1 or gpl20 stimulates cycles of cofilin phosphorylation and
  • cofilin is a major part of the motility engine in T cells
  • the T cell migratory defects seen in HIV-infected patients may directly result from cofilin dysregulation by persistent, pathogenic signaling occurring during HIV infection (50).
  • HIV infection during acute phase, gpl20 levels are very high with active HIV replication.
  • CD4 T cells are exposed to these high levels of gpl20 for extended periods of time before ART (Antiretroviral therapy) initiation.
  • ART Antiretroviral therapy
  • gpl20 can be present in high concentrations (>300 pg/ml), and is estimated to be in the range of 10 pg/ml to 10 ng/ml (51).
  • the CD4 T cells in HIV patients are chronically exposed to viral proteins such as gpl20 (52), particularly during the acute phase, persistent viral signaling may trigger cofilin dysregulation and cause a T cell migratory defect, as seen in HIV patients (2, 3, 50).
  • Fig. 2A blood resting CD4 T cells
  • hyperactivation may result from ART -irreversible, pathogenic polarization of T cells. This irreversibility appears to resemble the establishment of an early immune activation set point that dictates subsequent CD4 T cell depletion independent of viral load (1).
  • Both IR and INR had the viral load suppressed to the limit of detection after one year of treatment; the INR had less than 20% recovery of CD4 T cells or a CD4 T cell count below 200, whereas the IR had greater than 20% T cell recovery and a CD4 count above 500.
  • higher levels of p-cofilin in ART -treated patients were associated with a better CD4 T cell recovery after ART.
  • ART -naive patients were followed after their p-cofilin profiling. Some of these patients were subsequently treated with ART (Table 2). Again, the IR had significantly higher levels of cofilin phosphorylation than the INR (Fig. 2G).
  • Cofilin hyperactivation has been shown to be associated with a migratory impairment of CCR6+ and CXCR3+ helper T cells (Th), which are prevented from trafficking from the blood stream to peripheral organs even in aviremic HIV patients on long-term ART (2).
  • T cell migration is controlled by cycles of cofilin phosphorylation and dephosphorylation, which are regulated by the LIM-domain kinase (LIMK) through serine 3 phosphorylation (27, 54).
  • LIMK LIM-domain kinase
  • R10015 a recently discovered LIMK inhibitor
  • This HIV-mediated T cell polarization may resemble the irreversibility of T cell differentiation and lineage commitment following persistent cytokine receptor signaling.
  • T cells when T cells are stimulated with IL-12 or interferon-g for an extended period of time, they are polarized to express a transcription factor, T-bet, and the induction of which becomes irreversible.
  • the ART-irreversibility of cofilin hyperactivation also appears to resemble the establishment of the early immune activation set point that dictates subsequent CD4 T cell dysfunction and depletion independent of viral load (1).
  • the CD8 T cells cytotoxic T cells
  • monocytes carry higher-levels of cofilin phosphorylation
  • Fig. 5C and 5D cofilin dysregulation in T cells and other mononuclear cells of cancer patients, which can cause immune dysfunction and impair immune cell mobility.
  • Fig. 5A and B there is a narrowing of p-cofilin staining in the CD8 T cells of cancer patients (Fig. 5A and B), suggesting a decrease in T cell diversity, and a tumor-driving T cell lineage polarization in cancers, which can cause T cell dysfunction and impair T cell mobility.
  • HIV-1 infected patients were enrolled.
  • 98 had no previous or current ART at the time of the p-cofilin profiling, and 102 had ongoing ART for over a year, but 4 of the ART-treated patients had a viral load greater than 1,000 copies/ml and were excluded from the study for possible drug resistance.
  • the CD4 T cell count and viral load of these subjects were measured every 3 months.
  • One hundred age- and sex- matched healthy controls (HC) were enrolled.
  • a summary of the subjects is listed in Table 1 and Table 2.
  • 65 eventually received ART at around 6 months after the p-cofilin profiling, and were treated for more than a year.
  • ART-treated patients were further evaluated and categorized into immune responders (IR) and immune non-responders (INR). Both IR and INR were treated with ART for over one year. IRs were those who had a CD4 T cell recovery greater than 20% and a CD4 T cell count higher than 500 cells/ m ⁇ ; INRs had a CD4 T cell recovery less than 20% or a CD4 T cell count lower than 200 cells/ m ⁇ .
  • IR immune responders
  • INR immune non-responders
  • peripheral blood mononuclear cells were freshly obtained from the subjects and purified by Ficoll-Hypaque density gradient centrifugation, followed by negative isolation of resting CD4 T cells as previously described (9, 57). Briefly, monoclonal antibodies against human CD14, CD56, HLA-DR, CD8, CDllb, and CD 19 (BD Biosciences, San Jose, CA) were used. Antibody -bound cells were depleted using Dynabeads Pan Mouse IgG (Thermo Fisher Scientific). Purified cells were cultured in RPMI 1640 medium supplemented with 10% FBS.
  • the RPPA technology has been developed and optimized for performance as a fluorescence-based calibrated assay, generally identical in design and analysis to standard ELISA or standard clinical immunoassays.
  • Each array consists of patient cell lysates printed in triplicate two- spot dilutions (neat and 1 :4), high and low controls printed in triplicate two-spot dilutions (neat and 1 :4), and 6-10 point calibrators.
  • the analyte concentration is determined by extrapolation to a non-parametrically determined curve fit of the calibration curve and reported in relative fluorescence units.
  • each protein analyte value (relative p-cofilin value) is normalized to the total amount of protein printed on that spot with a fluorescent stain (Sypro Ruby Blot Stain, Molecular Probes, Eugene OR) that binds to the amne group of proteins without bias.
  • the protein loading value is also obtained by a calibrated assay technique.
  • This total protein calibrator consists of a protein lysate with a known concentration, which upon dilution spans the linear dynamic range of protein concentration. Each sample value is then extrapolated to the calibrator.
  • PBMC Peripheral blood mononuclear cells
  • Antibody-bound cells were depleted using Dynabeads Pan Mouse IgG (Invitrogen, Carlsbad, CA). For further negative selection of the memory and naive CD4 T cell subsets, monoclonal antibody against either CD45RA (0.02 m ⁇ per million cells) or CD45RO (0.1 m ⁇ per million cells) (BD
  • the blots were incubated with a mouse anti-cofilin antibody (1 :1000 dilution) (BD Biosciences, San Jose, CA) and a rabbit anti-phospho-cofilin (ser3) antibody (1 :500 dilution) (Cell Signaling) diluted in 3% milk- TBST and rocked overnight at 4°C. The blots were washed three times for 15 minutes, then incubated with DyLight 680 goat anti-mouse and DyLight 800 goat anti-rabbit antibodies (KPL, Gaithersburg, MD)
  • a half million cells were resuspended into 100 m ⁇ RPMI-1640 medium and then added to the upper chamber of a 24-well transwell plate (Corning, Coming, NY).
  • the lower chamber was filled with 600 m ⁇ of medium premixed with CXCL12 (40 ng/ml).
  • the plate was incubated at 37°C for 2 hours, and then the upper chamber was removed and cells in the lower chamber were counted.
  • Z2 Coulter Particle Count and Size Analyzer (Beckman Coulter) was used. Where indicated, different concentrations of R10015 (55) or DMSO were added to cell culture, incubated for 1 hour at 37°C before adding cells to the upper chamber.
  • Cells were also treated with the anti-human a4b7 integrin antibody (Act-1) or the 'control mouse IgGl antibody for 15 minutes before adding cells to the upper chamber. Act-1 was also added to the lower chamber (1 pg/ml) with CXCL12 (40 ng/ml). Multiple donors were used for chemotaxis assay.
  • One million cells were fixed, permeabilized with methanol, washed, and then stained with an anti-human p-cofilin antibody using an intracellular protein staining kit (Virongy, Manassas, VA) for 60 min at room temperature. Cells were washed twice and stained with Alexa Fluor 488-labeled chicken anti-rabbit antibodies (Invitrogen, Carlsbad, CA). Cells were washed twice, and then analyzed on a FACSCalibur (BD Biosciences, San Jose, CA).
  • Cecchinato V Bernasconi E, Speck RF, Proietti M, Sauermann U, D'Agostino G, Danelon G, Rezzonico Jost T, Grassi F, Raeli L, Schoni-Affolter F, Stahl-Hennig C,
  • HIV-1 entry cofactor functional cDNA cloning of a seven-transmembrane, G protein-coupled receptor. Science.
  • CC CKR5 a RANTES, MIP-1 alpha, MIP-lbeta receptor as a fusion cofactor for macrophage-tropic HIV- 1. Science. 1996;272(5270): 1955-8. PubMed PMID: 8658171.
  • Galkin VE Orlova A
  • VanLoock MS Shvetsov A
  • Reisler E Egelman EH.
  • ADF/cofilin use an intrinsic mode of F-actin instability to disrupt actin filaments. J Cell Biol. 2003; 163(5): 1057-66. PubMed PMID: 14657234.
  • PubMed PMID 11832213. 30. Gohla A, Birkenfeld J, Bokoch GM. Chronophin, a novel HAD-type serine protein phosphatase, regulates cofilin-dependent actin dynamics. Nat Cell Biol. 2005;7(l):21-9. PubMed PMID: 15580268.
  • HIV-1 envelope protein gpl20 is present at high concentrations in secondary lymphoid organs of individuals with chronic HIV- 1 infection. J Infect Dis. 2009;200(7): 1050-3. PubMed PMID: 19698075.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Biochemistry (AREA)
  • Cell Biology (AREA)
  • Virology (AREA)
  • Biotechnology (AREA)
  • Food Science & Technology (AREA)
  • Organic Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Microbiology (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Veterinary Medicine (AREA)
  • Hospice & Palliative Care (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • AIDS & HIV (AREA)
  • Toxicology (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Oncology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

A major immune dysfunction persisting in chronic diseases such as HIV infection and cancer is the impairment of T cell motility and migration to tissues. Therapeutics such as antiretroviral therapy or cancer therapy frequently do not fully restore T cell motility for tissue migration and infiltration. Cofilin is an actin-depolymerizing factor that regulates actin dynamics for T cell migration. Here we demonstrate that the levels of cofilin phosphorylation in blood T cells (CD4 or CD8), macrophages, B cells, natural killer (NK) cells, and/or cancer cells can be used to quantify the immune damages caused by chronic viral infection and cancer, and to predict the recovery of T cells following treatment.

Description

COFILIN PHOSPHORYLATION FOR CANCER TREATMENT
INTRODUCTION
Antiretroviral therapy (ART) has significantly extended the lifespan of HIV-infected people, but it offers neither a cure nor full immune restoration. The natural course of HIV infection leads to multiple CD4 T cell defects (1), including the impairment of T cell migration and homing to lymphoid tissues such as GALT (Gut-Associated Lymphoid Tissues) (2-4). Even with near-complete viral suppression with ART, normal levels of CD4 T cell repopulation to lymphoid tissues are rarely achieved (2, 3, 5, 6), thereby dampening immune responses and preventing full immune reconstitution for lasting viremia control (7). In HIV-infected patients, the vast majority of circulating CD4 T cells are not HIV-infected (0.2-16.4 HIV+ cells per million) (8). Thus, the T cell migratory defect seen in patients likely results from a bystander effect from chronic stimulation and receptor signaling by viral (9,
10) and/or inflammatory factors (1, 2). However, the molecular mechanism dictating this persistent T cell dysfunction had not been fully elucidated. This has hindered the
development of an effective therapy to restore T cell functions, and to achieve persistent immune control and clearance of HIV.
In cancers, there is also a similar impairment of T cell migration and infiltration into tumors (11). T cells such as CD8 T cells play a critical role in cancer immunity, but tumors can use various defense mechanisms, either directly or indirectly, to inhibit T migration to tumor bed for immune control (11).
In the human immune system, T cell activity is mainly regulated by receptor signaling. Persistent stimulation and signaling through chemotactic receptors (e.g., chemokine receptors, cytokine receptors, integrin receptors, adhesion molecule receptors, and co-stimulatory molecule receptors such as CD28, CTLA-4, and PD-l/PD-L) frequently leads to T cell polarization and commitment to distinct lineages. In HIV infection, the virus infects T cells through gpl20 binding to CD4 and the chemokine co-receptor CXCR4 (X4) or CCR5 (R5) (12, 13). Such binding also initiates aberrant signaling and has pathogenic consequences (9, 10, 14, 15). In particular, HIV signaling through CXCR4 has been shown to activate an actin depolymerizing factor, cofilin, to promote the actin dynamics necessary for viral nuclear entry in blood resting CD4 T cells (9, 16). Similarly, in other chronic diseases such as cancer, the persistent stimulation of immune cells by high levels of inflammatory chemokines, such as IP-10 (17, 18), and tumor antigens can also cause T polarization and dysfunction, and impair T cell motility (Fig. 1)
A major cellular protein controlling T cell motility and migration is cofilin. Cofilin is an actin-binding protein, one of the membranes of the ADF/cofilin family proteins ubiquitously present among eukaryotes. ADF/cofilin proteins bind and depolymerize filamentous F-actin in a pH-dependent manner and are responsible for the high turnover rates of actin filaments in cells. The first member of the ADF/cofilin family proteins was identified in extracts of embryonic chick brain (19). Cofilin was later purified from porcine brain and named cofilamentous structures with actin, or cofilin for short. Currently, there are three highly conserved proteins in the ADF/cofilin family: ADF (actin depolymerizing factor) or destrin, cofilin 1 (non-muscle cofilin or cofilin), and cofilin-2 (muscle-cofilin). Cloned human cofilin has 166 amino acids and a deduced molecular weight of 18 kD (20).
Structurally, human cofilin possesses a core consisting of a five-stranded mixed b- sheet (bΐ to b5). The first four strands (bΐ to b4) are anti-parallel, and b5 runs parallel to b4 and anti -parallel to b3. In addition, the residues 159-161 at the C-terminal form a short stand, b6. Five helices (al to a.5) surround the central b-sheet, and a salt bridge is formed between His133 and Asp98 that may influence the pH sensitivity of cofilin in actin binding and depolymerization. Cofilin binds to G actin, and the binding site is located in a region centered around Tyr117, which includes several residues in a.4 (such as Lys112, Lys114, Met115, and He124) and b5. In addition, a few other residues such as He12, Pro110 and Leu128 are also suggested in G-actin binding. Cofilin also binds to F-actin, and this requires two sites on cofilin, a G site that interacts with actin subdomains 1 and 3 and an F site that interacts with actin subdomains 1 and 2. Residues important for F-actin binding on cofilin have been identified through mutagenesis and include several residues in b4 (such as Lys96 and Asp98) and a.5 (such as Glu151, Lys152, and Gly155). Other regions including b3 and b5 may additionally be involved in F-actin binding (21).
Cofilin depolymerizes actin filaments through two mechanisms: direct severing and increasing the off-rate of actin subunits from the (-) end (22, 23). Cofilin has a higher affinity for ADP-actin than for ATP-actin, and cofilin binding to actin filaments facilitates phosphate dissociation from ATP-actin. Hydrolysis of ATP increases cofilin binding affinity, changing the twist of the actin helix and severing actin filaments to shorter segments (24). These severed actin filaments can increase the speed of actin polymerization by providing more free (+) ends for nucleation by the Arp2/3 complex (25).
The binding of cofilin to ADP-actin present at the (-) end also promotes dissociation of cofilin-ADP-actin. Cofilin in the released ADP-actin complex is competitively replaced by profilin, which then coverts ADP-actin monomers to ATP-actin monomers to recycle them for new actin polymerization at the (+) end. Thus, it is suggested that cofilin and the Arp2/3 complex work together to regulate actin treadmilling, in which ATP-actin is preferentially incorporated into the filaments through Arp2/3 at the (+) end, and then hydrolyzed into “older” ADP-actin and dissociated from the (-) end by cofilin (23, 26).
In cells, the activity of cofilin is mainly regulated by phosphorylation of serine 3 at the N-terminal, which inhibits cofilin binding to G-actin and F-actin. The kinases responsible for cofilin serine 3 phosphorylation are the LIM domain kinases (LIMK) and Tes kinases, which are targets of the Rho family GTPases such as Rho, Rac, and Cdc42 (27). Rho family GTPases activate PAK1, PAK2, PAK4, ROCK, or MRCKa (myotonic dystrophy kinase- related Cdc42-binding protein kinase) which then activate LIMK through direct
phosphorylation. Cofilin is activated through dephosphorylation of serine 3 by phosphatases such as PP1, PP2A, slingshot IL (SSH1L), and chronophin, which couple cofilin activity to different signal pathways (28-30). Recent studies have suggested that tyrosine 68 (Y68) can also be phosphorylated, and this phosphorylation appears to increase cofilin ubiquitination and proteasome degradation (31). In chemotactic cells, cycles of cofilin phosphorylation and dephosphorylation are required to sustain the actin dynamics essential for driving directional cell migration.
In cells, cofilin is a key regulator of actin dynamics and is also involved in multiple cellular processes (32). The ability of cofilin to modulate actin polymerization or
depolymerization may depend on the local concentration of cofilin. Low concentrations of cofilin favor severing, whereas high concentrations favor actin nucleation. Nevertheless, it is unknown how this concentration-dependent mode of action demonstrated in test tubes plays out in cells (33). In general, decreasing cofilin expression through siRNA increases the amounts of cellular actin filaments, whereas over-expression of cofilin induces the formation of cofilin-actin bundles (9). In vitro, cofilin also competitively interacts with the Arp2/3 complex by inducing structural changes in the actin filaments; these changes reduce the affinity for Arp2/3 and cause a loss of actin filament branches. This debranching process may play a role in modulating Arp2/3 -induced actin branch growth in the leading edge of migrating cells (34).
In addition to regulating actin dynamics in cells, cofilin has also been shown to mediate actin nuclear localization, which may be involved in the regulation of gene expression. Actin and actin-related proteins such as Arp7, Arp9, and Baf53 are parts of the chromatin-remodeling complex RSC and SWPSNF (35, 36). Actin is also part of the pre initiation complexes and is necessary for transcription by RNA polymerase II (37). Cofilin- mediated actin nuclear localization may serve to connect the cytoskeletal processes to chromatin remodeling and the initiation of transcription.
Cofilin serine 3 phosphorylation inactivates the actin binding ability of cofilin.
However, phospho-cofilin is recently shown to activate phospholipase D1 (PLD1). Cofilin directly and specifically interacts with PLD1 upon phosphorylation by LIMK1. Phospho- cofilin also stimulates PLD1 activity, suggesting that phospho-cofilin may control a variety of cellular functions by its stimulatory effect on PLD1 (38).
In the human immune system, cofilin plays important roles in regulating T cell migration, chemotaxis, and T cell activation. Within the immunological synapse, cofilin is required for the formation of the supramolecular activation clusters critical for sustaining signaling and T cell activation. Cell-permeable peptides that block cofilin interaction with F- actin impair receptor capping and immunological synapse formation, resulting in inhibition of T cell activation (39). In human blood resting T cells, in the absence of T cell activation or chemotactic stimulation, cofilin exists largely as the serine 3 phosphorylated form. T cell activation or chemotactic stimulation leads to transient cofilin activation by
dephosphorylation, and the signaling cascade is mainly transduced through costimulatory receptors such as CD2, CD28, and the chemokine receptors such as CXCR4. While
TCR/CD3 stimulation activates the Arp2/3 complex for actin polymerization, CD28- mediated costimulation triggers cofilin activation, which is required for dynamic actin reorganization and sustaining T cell signaling (40). The GTPase Ras and PI3K
(phosphatidylinositol-3 -kinase) signaling cascade is suggested to mainly regulate
dephosphorylation of cofilin in unstimulated human blood T cells (41). Inhibition of either MAPK/ERK kinase or PI3K blocks Ras-induced and costimulation-induced cofilin dephosphorylation, whereas transient expression of a dominant negative form of H-Ras inhibits PI3K activation and cofilin dephosphorylation (41). In HIV infection, the virus enters into cells through binding to CD4 and the chemokine coreceptor, CXCR4 or CCR5. During this entry process, HIV-1 binding to CXCR4 also triggers a transient course of cofilin phosphorylation and dephosphorylation to increase actin dynamics in resting CD4 T cells (9, 42). It is suggested that the cortical actin in resting T cells is relatively static in the absence of T cell activation or chemotactic stimulation. This lack of actin activity limits viral early processes such as entry, DNA synthesis, and nuclear migration (9). Cofilin increases cortical actin dynamics and actin treadmilling, facilitating viral intracellular migration towards the nucleus (9). Cofilin and actin-mediated HIV nuclear localization is suggested to be essential for the establishment of HIV-1 latency in resting CD4 T cells (9, 16). Slight inhibition of cofilin expression through siRNA knockdown increases cortical actin density, which leads to an increase in HIV DNA synthesis but a decrease in the amounts of HIV- 1 nuclear DNA and early transcripts (9). Induction of cofilin activity using a human cofilin-derived peptide (S3) carrying the N- terminal 16 residues competitively inhibits cofilin phosphorylation through LIMK1, and this induction enhances HIV latent infection of resting CD4 T cells. A pharmacological drug, staurosporine, is also shown to induce gradual cofilin activation that enhances HIV latent infection of resting CD4 T cells following a transient treatment during infection (9). In addition, pre-treatment of resting CD4 T cells with chemokines such as CCL19, CXCL9, CXCL10, and CCL20 lead to cofilin activation and changes in actin filaments which greatly promote HIV nuclear migration and DNA integration (16, 43). Interestingly, exposing cells to mechanical shear stress, such as infecting cells under conditions of low speed spinning or spinoculation, also triggers cofilin activation and actin dynamics, resulting in the
upregulation of CXCR4 and a great enhancement of HIV-1 DNA synthesis and nuclear migration (44).
HIV-l-mediated cofilin activation in resting CD4 T cells is shown to be through the God-dependent signaling from CXCR4; pertussis toxin (PTX), a bacterial toxin inhibiting G- protein-coupled receptors by the ADP-ribosylation of God, inhibits cofilin activation and HIV-1 latent infection of resting T cells (9). Cofilin can be phosphorylated by LIMK, and HIV-1 binding to blood CD4 T cells and macrophages triggers rapid activation of LIMK1, coincident with HIV-mediated early actin polymerization in T cells (42). It is suggested that HIV-1 hijacks LIMKl/cofilin activity to directly regulate actin and CXCR4 dynamics critical for viral entry, postentry DNA synthesis, and nuclear migration. Inhibition of LIMK1 activity through siRNA knockdown decreases filamentous actin and T cell chemotaxis towards SDF- 1. The decrease in cortical actin density also leads to an increase in CXCR4 internalization and surface recycling. Thus, LIMK-mediated early cortical actin polymerization may result in a temporary block to CXCR4 internalization, facilitating viral fusion and CXCR4 signaling. The LIMK1 knockdown cells also support lower viral entry, DNA synthesis, and nuclear migration. In addition, transient treatment of resting CD4 T cells with a pharmacological agent, okadaic acid, activates LIMK and promotes HIV latent infection of resting CD4 T cells. The signaling pathway that mediates LIMK activation by HIV-1 is identified as the Racl, PAK1/2 and LIMK pathway in blood resting T cells. The activation is likely triggered by gpl20 signaling through both CD4 and CXCR4, as well as from both Gori and Gaq (42).
In transformed cancer cells, HIV binding to CD4 and CXCR4 is also suggested to trigger filamin A activation which then leads to RhoA-ROCK-LIMK activation. This activity is proposed to permit F-actin reorganization for receptor clustering (45). HIV gpl20-mediated cofilin phosphorylation is also suggested to inhibit T cell chemotaxis towards SDF-1 (46).
In addition to HIV-1 gpl20, another HIV pathogenic factor, Nef, is also suggested to regulate cofilin activity (47). Over-expression of Nef in a human cancer Jurkat T cell inhibits SDF-1 -induced membrane ruffling, actin rearrangement, and cell migration towards
CXCL12, CCL3, and CCL19 (47, 48). Over-expression of Nef in hamster CHO cells also inhibits wounding-induced cofilin activation in a cell wound healing assay. It has been suggested that the Nef-PAK2 complex is involved in the phosphorylation of cofilin, although Nef does not appear to alter the activity of PAK2 in an in vitro cofilin phosphorylation assay. Functionally, Nef-mediated cofilin dysregulation may affect migratory behavior of infected T cells (47).
SUMMARY
Provided herein are methodology, compositions, kits, and the like for determining the levels of cofilin phosphorylation in blood T cells (CD4 or CD8), which can be used to quantify the immune damages caused by chronic viral infection and cancer, and to predict the recovery of T cells following treatment. Based on this determination of a correlation between cofilin phosphorylation and immune dysfunction, suitable patients may be treated with a composition that causes cofilin dysregulation and restores cofilin phosphorylation.
For instance, and in no way limiting, levels of cofilin phosphorylation in HIV-infected patients can be used as a marker to quantify immune dysfunction and damages by the virus. In another embodiment, levels of cofilin phosphorylation in HIV-infected patients can be used to measure drug effects on immune function (ART and other immune modulating drugs). Levels of cofilin phosphorylation in HIV-infected patients can be used to predict the recovery of T cell functions from drug treatment (ART and other immune modulating drugs).
In another embodiment, levels of coflin phosphorylation in cancer cells can be used to quantify alteration in cell motility, migration, tissue infiltration, and metastasis.
In another embodiment, levels of cofilin phosphorylation in T cells (CD4/CD8) in cancer patients can be used to predict immune dysfunction in cancer, and predict drugs effects in restoring anti-tumor immunity.
In some embodiments, a method for detecting and treating immune dysfunction in a patient comprises (a) quantifying cofilin phosphorylation levels in T cells (CD4/CD8), monocytes/macrophages, B cells, natural killer (NK) cells, and/or cancer cells of said patient, wherein said patient with immune dysfunction has lower or higher cofilin phosphorylation levels than a control healthy person or patient; and (b) administering to said patient an effective amount of a composition that corrects cofilin dysregulation and restores cofilin phosphorylation.
In some embodiments, the patient has a chronic disease causing immune dysfunction. In some embodiments, the said chronic disease is HIV infection or cancer.
In some embodiments, said immune dysfunction is aberrant T cell, macrophage, B cell, NK cell activation, impairment of T cell, monocyte/macrophage, B cell, NK cell motility and/or tissue migration.
In some embodiments, the composition is a cofilin phosphatase inhibitor or cofilin kinase activator. In some embodiments, the composition comprises okadaic acid.
In some embodiments, a method for determining immune dysfunction in a patient, comprises quantifying cofilin phosphorylation in T cells (CD4/CD8),
monocyte/macrophages, B cells, natural killer cells, and/or cancer cells in said patient.
In some embodiments, the patient is a cancer patient or HIV-infected patient.
In some embodiments, the method for treating cancer in a patient comprises (a) quantifying cofilin phosphorylation levels in T cells (CD4/CD8), monocyte/macrophages, B cells, natural killer cells, and/or cancer cells of said patient, wherein said patient has lower or higher cofilin phosphorylation levels than a control healthy person; and (b) administering to said patient an effective amount of a composition that restores cofilin phosphorylation.
In some embodiments, the composition is a cofilin phosphatase inhibitor or cofilin kinase activator or an antibody against chemotactic receptors that can trigger cofilin phosphorylation or dephosphorylation or a small molecule that can trigger cofilin
phosphorylation or dephosphorylation. In some embodiments, the composition comprises okadaic acid.
In some embodiments, the method is for treating a patient having cancer, including, for example, patients who do not have HIV or AIDS.
BRIEF DESCRIPTION OF THE IGI RES
Figure imgf000010_0001
FIGURE 1. Persistent stimulation of T cell surface receptors can lead to T cell polarization to different lineages. (A) Persistent and chronic stimulation of chemotactic receptors (e.g., chemokine receptors, cytokine receptors, integrin receptors, adhesion molecule receptors, and co-stimulatory molecule receptors, such as CD28/B7, B7-2, CTLA- 4/B7, B7-2, and PD-l/PD-L) can leads to T cell polarization and commitment to distinct lineages, such as an irreversible pathogenic lineage with chronic cofilin hyperactivation. (B) The signaling pathways involved in cofilin activation through the stimulation of receptors.
(C) The signaling pathways involved in cofilin activation through the stimulation of integrin receptors (modified from Juliano, 2002, Annu Rev Pharmacol Toxicol 42:283).
FIGURE 2. Cofilin hyperactivation in HIV infection. (A) Flowchart of the clinical study. (B) Development of the reverse-phase cofilin microarray for profiling cofilin phosphorylation. Synthetic peptides or cell lysates were serially diluted (1 : 1) and printed onto the microarray slides, which were then stained with antibodies against either total cofilin (right) or phospho-cofilin (left). P-cofilin-S3, a synthetic cofilin peptide with serine 3 phosphorylated; cofilin-S3, a similar peptide with no serine 3 phosphorylation. A431 or HeLa cells were not treated or treated with human epithelial growth factor (EGF) or pervanadate (Perv). (C) Relative levels of p-cofilin in blood resting CD4 T cells from HIV-infected patients with ART (HIV+ART) or without ART (HIV), or healthy control donors (HC) were profiled. Box plots show interquartile range, median, and range. There were no statistically significant differences in the total protein levels of the resting CD4 T cells from HC, HIV, and HIV+ART (see Materials and Methods). (D and E) The correlation between levels of p- cofilin and plasma viral load (D) and CD4 T cell count (E) in untreated patients were plotted using Spearman rank correlation tests (Ln, natural logarithm). (F) In ART -treated patients, immune responders (IR) had significantly higher levels of cofilin phosphorylation than did non-responders (INR). (G) A subgroup of ART-naive patients was subsequently treated with ART following p-cofilin profiling. Immune responders (IR) had significantly higher levels of cofilin phosphorylation than non-responders (INR).
FIGURE 3. Quantification of effects of cofilin hyperactivation on T cell migration. (A) A3R5.7 T cells were treated with different dosages of R10015 for one hour. Phospho-cofilin and total cofilin were quantified by Western blot. (B) The relative ratio of p- cofilin/cofilin in response to R10015 treatment was plotted (n = 4 independent experiments). (C) R10015 inhibits cofilin phosphorylation and T cell chemotaxis in response to CXCL12. A3R5.7 cells were treated with different dosages of R10015 for one hour, and then added to the upper chamber of a 24-well transwell plate. The lower chamber was filled with CXCL12 (40 ng/ml), and cell migration to the lower chamber was quantified (n = 3 independent experiments). (D) The linear correlation between T cell migration and levels of cofilin phosphorylation. The X-axis is the relative ratio of p-cofilin/cofilin derived from (B); the Y- axis is the number of migrating cells derived from (C).
FIGURE 4. Cofilin hyperactivation in blood cancer cells. Peripheral blood mononuclear cells (PBMC) from healthy donors, Jurkat cancer T lymphocyte cells (derived from a patient with acute T cell leukemia), A3R5.7 T cells (derived from the peripheral blood huffy coat of a four-year-old patient with acute lymphoblastic leukemia), or THP-1 blood monocytic cancer cells (derived from an acute monocytic leukemia patient) were fixed, permeabilized, washed, and then stained with a rabbit polyclonal anti-human p-cofilin antibody for 60 min. Cells were washed twice and stained with Alexa Fluor 488-labeled anti rabbit antibodies. Cells were washed twice, and then analyzed by flow cytometry. The relative p-cofilin staining was calculated by using the mean fluorescent intensity from p- cofilin staining subtracting the mean fluorescent intensity from the non-specific background staining from similarly-labeled isotype control antibodies.
FIGURE 5: Cofilin dysregulation in blood CD8 T cells and monocytes of cancer patients. Peripheral blood cytotoxic T cells (CD8 T cells) or monocytes from health donors or colorectal cancer patients were fixed, permeabilized, washed, and then stained with a rabbit polyclonal anti-human p-cofilin antibody for 60 min. Cells were washed twice and stained with Alexa Fluor 488-labeled anti-rabbit antibodies. Cells were washed twice, and then analyzed by flow cytometry. The relative p-cofilin staining was calculated. (A and B) shows the representative p-cofilin staining from 8 donors. The histogram plots of p-cofilin staining in CD8 T cells from 2 healthy donors (A) or from 2 colorectal cancer patients (B). The filled grey plots are isotype staining, the red line plots are p-cofilin staining. As shown, in cancer patients there is a narrowing of p-confilin staining, suggesting a decrease of CD8 T cell diversity and a CD8 T cell lineage polarization, which can cause T cell dysfunction and affects T cell mobility. (C) The relative P-cofilin staining was calculated by using the mean fluorescent intensity from p-cofilin staining. As shown, CD8 T cells from cancer patients (n = 4 patients) have higher levels of p-cofilin staining than healthy donors (n = 4 donors), an indication of cofilin inactivation. (D) P-cofilin was similarly analyzed in blood monocytes of cancer patients and healthy donors.
FIGURE 6. Characteristics of clinical study participants. Details are in examples.
FIGURE 7. Patient enrollment and grouping. Details are in examples.
DETATUED DESCRIPTION
A major immune dysfunction persisting in chronic diseases such as HIV infection and cancer is the impairment of T cell motility and migration to tissues. Therapeutics such as antiretroviral therapy or cancer therapy frequently do not fully restore T cell motility for tissue migration and infiltration. Cofilin is an actin-depolymerizing factor that regulates actin dynamics for T cell migration.
As explained below, the present inventors determined that the levels of cofilin phosphorylation in blood T cells (CD4 or CD8) can be used to quantify the immune damages caused by chronic viral infection and cancer, and to predict the recovery of T cells following treatment. Based on this determination of a correlation between cofilin phosphorylation and immune dysfunction, suitable patients may be treated with a composition that corrects cofilin dysregulation and restores cofilin phosphorylation. For example, cells can be treated with cofilin phosphatase inhibitors or cofilin kinase activator to increase cofilin phosphorylation, as demonstrated by the use of okadaic acid to increase cofilin phosphorylation (42) or an anti- integrin antibody to modulate the cofilin pathway (49).
A potential pathogenic role of HIV- 1 -mediated cofilin dysregulation is proposed from studies of cofilin activation in blood resting CD4 T cells treated with HIV or gpl20 (50). It was shown that HIV-1 or gpl20 stimulates cycles of cofilin phosphorylation and
dephosphorylation, suggesting that chronic exposure of CD4 T cells to HIV or gpl20 may have a lasting impact on cofilin activity and T cell functionality. A small-scale clinical study has found that in the peripheral blood of HIV- 1 -infected patients, levels of active cofilin in their resting CD4 T cells are significantly higher. It is suggested that HIV- 1 -mediated dysregulation of cofilin may lead to abnormalities in T cell migration and activation that could contribute to viral pathogenesis (50).
Given that cofilin is a major part of the motility engine in T cells, it is possible that the T cell migratory defects seen in HIV-infected patients may directly result from cofilin dysregulation by persistent, pathogenic signaling occurring during HIV infection (50). In HIV infection, during acute phase, gpl20 levels are very high with active HIV replication. CD4 T cells are exposed to these high levels of gpl20 for extended periods of time before ART (Antiretroviral therapy) initiation. During the asymptomatic phase with ART, gpl20 in the peripheral blood is at a low level. However, in infected lymphoid tissues, gpl20 can be present in high concentrations (>300 pg/ml), and is estimated to be in the range of 10 pg/ml to 10 ng/ml (51). Given that the CD4 T cells in HIV patients are chronically exposed to viral proteins such as gpl20 (52), particularly during the acute phase, persistent viral signaling may trigger cofilin dysregulation and cause a T cell migratory defect, as seen in HIV patients (2, 3, 50).
A. Effects of HIV Infection on Cofilin Activity
To detect the effects of HIV infection on cofilin activity, and as explained below, the present inventors conducted a large clinical trial to examine cofilin phosphorylation in blood resting CD4 T cells (Fig. 2A). For this purpose, a reverse phase phospho-cofilin micro-array was developed that can quantify cofilin phosphorylation in a large number of clinical samples simultaneously (53) (Fig. 2B). Blood resting CD4 T cells from HIV patients, with ART (HIV+ART, n = 95) or without ART (HIV, n = 98), or from healthy controls (HC, n = 100) (Table 1) were purified by negative depletion, unstimulated, and then lysed. Blindly coded cell lysates were then profiled with the phospho-cofilin micro-array (Fig. 2C). As shown in the figures, the inventors observed a highly significant reduction in cofilin phosphorylation in HIV patients (HIV = 0.968; HIV+ART = 1.139; healthy control = 2.254; p< 0.001).
Surprisingly, ART did not significantly restore cofilin phosphorylation (HIV = 0.968;
HIV+ART = 1.139; p= 0.981). These results suggest that HIV-mediated cofilin
hyperactivation may result from ART -irreversible, pathogenic polarization of T cells. This irreversibility appears to resemble the establishment of an early immune activation set point that dictates subsequent CD4 T cell depletion independent of viral load (1). B. Cofilin hyperactivation and viral load/CD4 count
The inventors next examined possible correlations between cofilin hyperactivation and viral load/CD4 count. In untreated patients, there was only a weak correlation between cofilin phosphorylation and viral load (p = 0.043, r = -0.205) (Fig. 2D), and there was no correlation between cofilin phosphorylation and CD4 T cell counts (p = 0.057, r = 0.193) (Fig. 2E). However, when ART -treated patients were categorized into immune responders (IR) and immune non-responders (INR), the IR had a significantly higher level of cofilin phosphorylation than the INR (Fig. 2F). Both IR and INR had the viral load suppressed to the limit of detection after one year of treatment; the INR had less than 20% recovery of CD4 T cells or a CD4 T cell count below 200, whereas the IR had greater than 20% T cell recovery and a CD4 count above 500. Thus, higher levels of p-cofilin in ART -treated patients were associated with a better CD4 T cell recovery after ART. ART -naive patients were followed after their p-cofilin profiling. Some of these patients were subsequently treated with ART (Table 2). Again, the IR had significantly higher levels of cofilin phosphorylation than the INR (Fig. 2G). These results demonstrate that pre-ART levels of p-cofilin can be used to gauge the degree of CD4 T cell damage and predict T cell recovery from ART.
Cofilin hyperactivation has been shown to be associated with a migratory impairment of CCR6+ and CXCR3+ helper T cells (Th), which are prevented from trafficking from the blood stream to peripheral organs even in aviremic HIV patients on long-term ART (2). T cell migration is controlled by cycles of cofilin phosphorylation and dephosphorylation, which are regulated by the LIM-domain kinase (LIMK) through serine 3 phosphorylation (27, 54). To quantify the direct effect of cofilin hyperactivation on T cell migration, we used a recently discovered LIMK inhibitor, R10015 (55), to block cofilin phosphorylation in A3R5.7 CD4 T cells. We then performed CXCL12-induced T cell chemotaxis in the presence of cofilin hyperactivation. CXCL12 (SDF-1, stromal cell-derived factor 1) binding to CXCR4 triggers actin/cofilin dynamics for T cell migration (9). We observed a R10015 dosage- dependent inhibition of cofilin phosphorylation in A3R5.7 CD4 T cells (Fig. 3A and 3B). A linear correlation was observed between cofilin dephosphorylation and the impairment of CXCL12-mediated T cell chemotaxis (Fig. 3C and 3D) (correlation coefficient r = 0.999, p =0.002). At around 15 mM of R10015, cofilin phosphorylation was reduced to around 50% in A3R5.7, a level approximate to what was seen in HIV patients (Fig. 3A and Fig. 2C). A 50% reduction in cofilin phosphorylation resulted in a 20-40% decrease in cell migration for human A3R5.7 CD4 T cells (Fig. 3C). These results quantitatively measured the direct impact of cofilin hyperactivation on T cell motility.
It has long been speculated that HIV binding to chemokine coreceptors may trigger aberrant G protein signaling and CD4 T cell pathogenesis (12). Nevertheless, this speculation has not been solidified by experimental and clinical data. In this large clinical trial, the inventors demonstrated cofilin dephosphorylation occurring in the blood CD4 T cells of HIV- infected patients. Cofilin has been identified as a direct downstream target of HIV-mediated G protein signaling through the chemokine coreeptors (9). Cofilin hyperactivation can directly affect T cell migration (40), as cofilin is a major driver of actin treadmilling for cell motility (56). In HIV infection, selective impairment of CD4 but not CD8 T cell homing (2,
4) is a major hallmark of HIV-mediated T cell dysfunction (3), and this implies a direct role of cofilin dysregulation in HIV-mediated CD4 T cell pathogenesis. The molecular cue for this cofilin hyperactivation likely results from early viral signaling from HIV gpl20 (9) and chronic immune activation later in the disease course (2). It has been known that in HIV- infected patients, levels of inflammatory cytokines such as IP-10 are very high in the peripheral blood (17, 18). Thus, the combined effects of gpl20 and chronic immune activation may exacerbate and polarize CD4 T cells towards an ART-irreversible pathogenic lineage. This HIV-mediated T cell polarization may resemble the irreversibility of T cell differentiation and lineage commitment following persistent cytokine receptor signaling. For example, when T cells are stimulated with IL-12 or interferon-g for an extended period of time, they are polarized to express a transcription factor, T-bet, and the induction of which becomes irreversible. The ART-irreversibility of cofilin hyperactivation also appears to resemble the establishment of the early immune activation set point that dictates subsequent CD4 T cell dysfunction and depletion independent of viral load (1).
It was also found that similar to HIV- 1 -mediated cofilin hyperactivation, human cancer cells also carry lower-levels of cofilin phosphorylation (Fig. 4), demonstrating the motility of transformed cancer cells have been fundamentally altered through changing cofilin phosphorylation.
It was also found that in cancer patients, the CD8 T cells (cytotoxic T cells) and monocytes carry higher-levels of cofilin phosphorylation (Fig. 5C and 5D), demonstrating that there is a cofilin dysregulation in T cells and other mononuclear cells of cancer patients, which can cause immune dysfunction and impair immune cell mobility. In addition, there is a narrowing of p-cofilin staining in the CD8 T cells of cancer patients (Fig. 5A and B), suggesting a decrease in T cell diversity, and a tumor-driving T cell lineage polarization in cancers, which can cause T cell dysfunction and impair T cell mobility.
EXAMPLE
The following example is illustrative and non-limiting.
Example 1: Clinical study
200 HIV-1 infected patients were enrolled. Among the HIV-infected patients, 98 had no previous or current ART at the time of the p-cofilin profiling, and 102 had ongoing ART for over a year, but 4 of the ART-treated patients had a viral load greater than 1,000 copies/ml and were excluded from the study for possible drug resistance. The CD4 T cell count and viral load of these subjects were measured every 3 months. One hundred age- and sex- matched healthy controls (HC) were enrolled. A summary of the subjects is listed in Table 1 and Table 2. Of the ART-naive patients, 65 eventually received ART at around 6 months after the p-cofilin profiling, and were treated for more than a year. All of these patients receiving ART reached undetectable plasma HIV-1 RNA. ART-treated patients were further evaluated and categorized into immune responders (IR) and immune non-responders (INR). Both IR and INR were treated with ART for over one year. IRs were those who had a CD4 T cell recovery greater than 20% and a CD4 T cell count higher than 500 cells/ mΐ; INRs had a CD4 T cell recovery less than 20% or a CD4 T cell count lower than 200 cells/ mΐ. For isolating blood resting CD4 T cells from study subjects, peripheral blood mononuclear cells were freshly obtained from the subjects and purified by Ficoll-Hypaque density gradient centrifugation, followed by negative isolation of resting CD4 T cells as previously described (9, 57). Briefly, monoclonal antibodies against human CD14, CD56, HLA-DR, CD8, CDllb, and CD 19 (BD Biosciences, San Jose, CA) were used. Antibody -bound cells were depleted using Dynabeads Pan Mouse IgG (Thermo Fisher Scientific). Purified cells were cultured in RPMI 1640 medium supplemented with 10% FBS. One million resting CD4 T cells from each blood donor were lysed in 40 mΐ SDS/T-PER extraction buffer [Novex Tris-Glycine SDS Sample Buffer, T-PER Tissue Protein Extraction Reagent (Thermo Fisher Scientific) and 2.5% 2-mercaptoethanol (Sigma- Aldrich)]. Cell lysates were heated at 100°C for 8 minutes, immediately frozen and stored at -80°C, and then transported on dry ice to Theranostics Health (Gaithersburg, MD, USA) for p-cofilin reverse phase protein microarray analyses. A total of 296 coded cell lysates were printed onto the microarrays and profiled; 3 lysates did not generate readable signals and were excluded from data analyses. Reverse phase protein microarray (RPPA)
Cofilin reverse phase protein microarray printing and analyses of cell lysates were provided by Theranostics Health (Gaithersburg, MD, USA). Details of RPPA have been published previously (53). The RPPA directly couples the phospho-cofilin detection antibody with highly sensitive amplification systems that can yield detection sensitivities to fewer than 1,000 to 5,000 molecules per spot with good linearity (correlation coefficient or R2 = 0.990- 0.999) and inter-experiment precision (R2= 0.973). Published between-run and within-run analytical precision in our studies is a 3-13% CV (coefficient of variation) (58). The RPPA technology has been developed and optimized for performance as a fluorescence-based calibrated assay, generally identical in design and analysis to standard ELISA or standard clinical immunoassays. Each array consists of patient cell lysates printed in triplicate two- spot dilutions (neat and 1 :4), high and low controls printed in triplicate two-spot dilutions (neat and 1 :4), and 6-10 point calibrators. The analyte concentration is determined by extrapolation to a non-parametrically determined curve fit of the calibration curve and reported in relative fluorescence units. For data normalization, each protein analyte value (relative p-cofilin value) is normalized to the total amount of protein printed on that spot with a fluorescent stain (Sypro Ruby Blot Stain, Molecular Probes, Eugene OR) that binds to the amne group of proteins without bias. The protein loading value is also obtained by a calibrated assay technique. This total protein calibrator consists of a protein lysate with a known concentration, which upon dilution spans the linear dynamic range of protein concentration. Each sample value is then extrapolated to the calibrator. The quantified averages of the total protein levels from each test group are: HIV (n = 98), 0.254; HIV+ART (n = 95), 0.259; HC (healthy control, n =100), 0.252. There are no statistically significant differences in the total protein levels between the 3 groups (HC and HIV, p = 0.77; HC and HIV+ART p = 0.51; HIV and HIV+ART, p = 0.64).
Purification of resting CD4 T subtypes from peripheral blood
Peripheral blood mononuclear cells (PBMC) were purified from peripheral blood of HIV-negative donors by centrifugation in Lymphocyte Separation Medium (Corning, Corning, NY), and resting CD4 T cells were further purified by two rounds of negative selection as previously described (9, 57). Briefly, for the first-round depletion, monoclonal antibodies against human CD14, CD56 and HLA-DR, DP, and DQ (BD Biosciences, San Jose, CA) were used. For the second-round depletion, monoclonal antibodies against human CD8, CD1 lb, and CD 19 (BD Biosciences, San Jose, CA) were used. Antibody-bound cells were depleted using Dynabeads Pan Mouse IgG (Invitrogen, Carlsbad, CA). For further negative selection of the memory and naive CD4 T cell subsets, monoclonal antibody against either CD45RA (0.02 mΐ per million cells) or CD45RO (0.1 mΐ per million cells) (BD
Biosciences, San Jose, CA) was added during the second round of depletion. Purified cells were cultured in RPMI-1640 medium supplemented with 10% heat-inactivated fetal bovine serum (Invitrogen, Carlsbad, CA), penicillin (50 U/ml) (Invitrogen, Carlsbad, CA), and streptomycin (50 pg/ml) (Invitrogen, Carlsbad, CA). Cells were rested overnight before infection or treatment. For a4b7 surface receptor upregulation, resting CD4 T cells were also cultured in IL-7 (5 ng/ml) for 3 days.
Western blotting for p-cofilin and cofilin
One million cells were lysed in NuPAGE LDS Sample Buffer (Invitrogen, Carlsbad, CA) followed by sonication. Samples were heated at 70°C for 10 minutes, separated by SDS- PAGE, and then transferred onto nitrocellulose membranes (Invitrogen, Carlsbad, CA). The membranes were washed in TBST for 3 minutes and then blocked for 30 minutes at room temperature with 5% milk. The blots were incubated with a mouse anti-cofilin antibody (1 :1000 dilution) (BD Biosciences, San Jose, CA) and a rabbit anti-phospho-cofilin (ser3) antibody (1 :500 dilution) (Cell Signaling) diluted in 3% milk- TBST and rocked overnight at 4°C. The blots were washed three times for 15 minutes, then incubated with DyLight 680 goat anti-mouse and DyLight 800 goat anti-rabbit antibodies (KPL, Gaithersburg, MD)
(1 :5000 diluted in blocking buffer) for 1 hour at 4°C. The blots were washed three times for 15 minutes and scanned with Odyssey Infrared Imager (Li-cor Biosciences).
Chemotaxis assay
A half million cells were resuspended into 100 mΐ RPMI-1640 medium and then added to the upper chamber of a 24-well transwell plate (Corning, Coming, NY). The lower chamber was filled with 600 mΐ of medium premixed with CXCL12 (40 ng/ml). The plate was incubated at 37°C for 2 hours, and then the upper chamber was removed and cells in the lower chamber were counted. To ensure accurate enumeration of cells, only Z2 Coulter Particle Count and Size Analyzer (Beckman Coulter) was used. Where indicated, different concentrations of R10015 (55) or DMSO were added to cell culture, incubated for 1 hour at 37°C before adding cells to the upper chamber. Cells were also treated with the anti-human a4b7 integrin antibody (Act-1) or the 'control mouse IgGl antibody for 15 minutes before adding cells to the upper chamber. Act-1 was also added to the lower chamber (1 pg/ml) with CXCL12 (40 ng/ml). Multiple donors were used for chemotaxis assay.
Intracellular p-cofilin staining and flow cytometry
One million cells were fixed, permeabilized with methanol, washed, and then stained with an anti-human p-cofilin antibody using an intracellular protein staining kit (Virongy, Manassas, VA) for 60 min at room temperature. Cells were washed twice and stained with Alexa Fluor 488-labeled chicken anti-rabbit antibodies (Invitrogen, Carlsbad, CA). Cells were washed twice, and then analyzed on a FACSCalibur (BD Biosciences, San Jose, CA).
Statistical analysis
Statistical calculations were performed using IBM SPSS statistics 23. Categorical data were described and analyzed by frequency and chi-square (c2) test. For parametric comparison, two-tailed Mann-Whitney U test was used to assess differences between groups during p-cofilin profiling (Fig. 2). Spearman rank correlations tests were used to measure the correlations between variables. Unless otherwise stated, p value less than 0.05 was considered as statistically significant.
References
1. Deeks SG, Kitchen CM, Liu L, Guo H, Gascon R, Narvaez AB, Hunt P, Martin JN, Kahn JO, Levy J, McGrath MS, Hecht FM. Immune activation set point during early HIV infection predicts subsequent CD4+ T-cell changes independent of viral load. Blood.
2004; 104(4) : 942-7. PubMed PMID: 15117761.
2. Cecchinato V, Bernasconi E, Speck RF, Proietti M, Sauermann U, D'Agostino G, Danelon G, Rezzonico Jost T, Grassi F, Raeli L, Schoni-Affolter F, Stahl-Hennig C,
Uguccioni M, Swiss HIVCS. Impairment of CCR6+ and CXCR3+ Th Cell Migration in HIV- 1 Infection Is Rescued by Modulating Actin Polymerization. J Immunol. 2017; 198(1): 184-95. doi: 10.4049/jimmunol.1600568. PubMed PMID: 27895171; PMCID: PMC5164881.
3. Mavigner M, Cazabat M, Dubois M, L'Faqihi FE, Requena M, Pasquier C, Klopp P, Amar J, Alric L, Barange K, Vinel JP, Marchou B, Massip P, Izopet J, Delobel P. Altered CD4+ T cell homing to the gut impairs mucosal immune reconstitution in treated HIV- infected individuals. J Clin Invest. 2012; 122(l):62-9. doi: 10.1172/JCI59011. PubMed PMID: 22156200; PMCID: PMC3248296. 4. Perez -Patrigeon S, Vingert B, Lambotte O, Viard JP, Delfraissy JF, Theze J,
Chakrabarti LA. HIV infection impairs CCR7-dependent T-cell chemotaxis independent of CCR7 expression. Aids. 2009;23(10): 1197-207. PubMed PMID: 19455014.
5. Ryan ES, Micci L, Fromentin R, Paganini S, McGary CS, Easley K, Chomont N, Paiardini M. Loss of Function of Intestinal IL-17 and IL-22 Producing Cells Contributes to Inflammation and Viral Persistence in SIV-Infected Rhesus Macaques. PLoS Pathog.
2016; 12(2):el 005412. doi: 10.1371/journal.ppat.1005412. PubMed PMID: 26829644;
PMC ID: PMC4735119.
6. Cecchinato V, Trindade CJ, Laurence A, Heraud JM, Brenchley JM, Ferrari MG, Zaffiri L, Tryniszewska E, Tsai WP, Vaccari M, Parks RW, Venzon D, Douek DC, O'Shea JJ, Franchini G. Altered balance between Thl7 and Thl cells at mucosal sites predicts AIDS progression in simian immunodeficiency virus-infected macaques. Mucosal Immunol.
2008;l(4):279-88. doi: 10.1038/mi.2008.14. PubMed PMID: 19079189; PMCID:
PMC2997489.
7. Byrareddy SN, Arthos J, Cicala C, Villinger F, Ortiz KT, Little D, Sidell N, Kane MA, Yu J, Jones JW, Santangelo PJ, Zurla C, McKinnon LR, Arnold KB, Woody CE, Walter L, Roos C, Noll A, Van Ryk D, Jelicic K, Cimbro R, Gumber S, Reid MD, Adsay V, Amancha PK, Mayne AE, Parslow TG, Fauci AS, Ansari AA. Sustained virologic control in SIV+ macaques after antiretroviral and alpha4beta7 antibody therapy. Science.
2016;354(6309): 197-202. doi: 10.1126/science.aagl276. PubMed PMID: 27738167;
PMCID: PMC5405455.
8. Finzi D, Hermankova M, Pierson T, Carruth LM, Buck C, Chaisson RE, Quinn TC, Chadwick K, Margolick J, Brookmeyer R, Gallant J, Markowitz M, Ho DD, Richman DD, Siliciano RF. Identification of a reservoir for HIV-1 in patients on highly active antiretroviral therapy. Science. 1997;278(5341): 1295-300.
9. Yoder A, Yu D, Dong L, Iyer SR, Xu X, Kelly J, Liu J, Wang W, Vorster PJ, Agulto L, Stephany DA, Cooper JN, Marsh JW, Wu Y. HIV envelope-CXCR4 signaling activates cofilin to overcome cortical actin restriction in resting CD4 T cells. Cell. 2008;134(5):782- 92. PubMed PMID: 18775311.
10. Wu Y, Yoder A. Chemokine coreceptor signaling in HIV-1 infection and
pathogenesis. PLoS Pathog. 2009;5(12):el000520. PubMed PMID: 20041213. 11. Lanitis E, Dangaj D, Irving M, Coukos G. Mechanisms regulating T-cell infiltration and activity in solid tumors. Ann Oncol. 2017;28(suppl_12):xiil8-xii32. doi:
10.1093/annonc/mdx238. PubMed PMID: 29045511.
12. Feng Y, Broder CC, Kennedy PE, Berger EA. HIV-1 entry cofactor: functional cDNA cloning of a seven-transmembrane, G protein-coupled receptor. Science.
1996;272(5263): 872-7. PubMed PMID: 8629022.
13. Alkhatib G, Combadiere C, Broder CC, Feng Y, Kennedy PE, Murphy PM, Berger EA. CC CKR5: a RANTES, MIP-1 alpha, MIP-lbeta receptor as a fusion cofactor for macrophage-tropic HIV- 1. Science. 1996;272(5270): 1955-8. PubMed PMID: 8658171.
14. Weissman D, Rabin RL, Arthos J, Rubbert A, Dybul M, Swofford R, Venkatesan S, Farber JM, Fauci AS. Macrophage-tropic HIV and SIV envelope proteins induce a signal through the CCR5 chemokine receptor. Nature. 1997;389(6654):981-5.
15. Schweneker M, Favre D, Martin JN, Deeks SG, McCune JM. HIV-induced changes in T cell signaling pathways. J Immunol. 2008;180(10):6490-500. PubMed PMID: 18453567; PMC ID: PMC2648824.
16. Cameron PU, Saleh S, Sallmann G, Solomon A, Wightman F, Evans VA, Boucher G, Haddad EK, Sekaly RP, Harman AN, Anderson JL, Jones KL, Mak J, Cunningham AL, Jaworowski A, Lewin SR. Establishment of HIV-1 latency in resting CD4+ T cells depends on chemokine-induced changes in the actin cytoskeleton. Proc Natl Acad Sci U S A.
2010; 107(39): 16934-9. PubMed PMID: 20837531.
17. Wang Z, Shang H, Jiang Y. Chemokines and Chemokine Receptors: Accomplices for Human Immunodeficiency Virus Infection and Latency. Front Immunol. 2017;8: 1274. doi: 10.3389/fimmu.2017.01274. PubMed PMID: 29085362; PMCID: PMC5650658.
18. Wang Z, Wu T, Ma M, Zhang Z, Fu Y, Liu J, Xu J, Ding H, Han X, Chu Z, Wu Y, Shang H, Jiang Y. Elevated interferon-gamma-induced protein 10 and its receptor CXCR3 impair NK cell function during HIV infection. J Leukoc Biol. 2017; 102(1): 163-70. doi:
10.1189/jlb.5A1016-444R. PubMed PMID: 28465448.
19. Bamburg JR, Harris HE, Weeds AG. Partial purification and characterization of an actin depolymerizing factor from brain. FEBS Lett. 1980; 121(1): 178-82. PubMed PMID: 6893966. 20. Bamburg JR. Proteins of the ADF/cofilin family: essential regulators of actin dynamics. Annu Rev Cell Dev Biol. 1999;15: 185-230. PubMed PMID: 10611961.
21. Pope BJ, Zierler-Gould KM, Kuhne R, Weeds AG, Ball LJ. Solution structure of human cofilin: actin binding, pH sensitivity, and relationship to actin-depolymerizing factor. J Biol Chem. 2004;279(6):4840-8. PubMed PMID: 14627701.
22. Pavlov D, Muhlrad A, Cooper J, Wear M, Reisler E. Actin filament severing by cofilin. J Mol Biol. 2007;365(5): 1350-8. PubMed PMID: 17134718.
23. Carlier MF, Laurent V, Santolini J, Melki R, Didry D, Xia GX, Hong Y, Chua NH, Pantaloni D. Actin depolymerizing factor (ADF/cofilin) enhances the rate of filament turnover: implication in actin-based motility. J Cell Biol. 1997; 136(6): 1307-22. PubMed PMID: 9087445.
24. Galkin VE, Orlova A, VanLoock MS, Shvetsov A, Reisler E, Egelman EH.
ADF/cofilin use an intrinsic mode of F-actin instability to disrupt actin filaments. J Cell Biol. 2003; 163(5): 1057-66. PubMed PMID: 14657234.
25. Pollard TD, Borisy GG. Cellular motility driven by assembly and disassembly of actin filaments. Cell. 2003;112(4):453-65. PubMed PMID: 12600310.
26. McGough A, Pope B, Chiu W, Weeds A. Cofilin changes the twist of F-actin:
implications for actin filament dynamics and cellular function. J Cell Biol. 1997;138(4):771- 81. PubMed PMID: 9265645.
27. Yang N, Higuchi O, Ohashi K, Nagata K, Wada A, Kangawa K, Nishida E, Mizuno
K. Cofilin phosphorylation by LIM-kinase 1 and its role in Rac-mediated actin
reorganization. Nature. 1998;393(6687):809-12. PubMed PMID: 9655398.
28. Ambach A, Saunus J, Konstandin M, Wesselborg S, Meuer SC, Samstag Y. The serine phosphatases PP1 and PP2A associate with and activate the actin-binding protein cofilin in human T lymphocytes. Eur J Immunol. 2000;30(12):3422-31. PubMed PMID: 11093160.
29. Niwa R, Nagata-Ohashi K, Takeichi M, Mizuno K, Uemura T. Control of actin reorganization by Slingshot, a family of phosphatases that dephosphorylate ADF/cofilin.
Cell. 2002;108(2):233-46. PubMed PMID: 11832213. 30. Gohla A, Birkenfeld J, Bokoch GM. Chronophin, a novel HAD-type serine protein phosphatase, regulates cofilin-dependent actin dynamics. Nat Cell Biol. 2005;7(l):21-9. PubMed PMID: 15580268.
31. Yoo Y, Ho HJ, Wang C, Guan JL. Tyrosine phosphorylation of cofilin at Y68 by v- Src leads to its degradation through ubiquitin-proteasome pathway. Oncogene.29(2):263 -72. PubMed PMID: 19802004.
32. Bernstein BW, Bamburg JR. ADF/cofilin: a functional node in cell biology. Trends Cell Biol. 2010;20(4): 187-95. PubMed PMID: 20133134.
33. Andrianantoandro E, Pollard TD. Mechanism of actin filament turnover by severing and nucleation at different concentrations of ADF/cofilin. Mol Cell. 2006;24(1): 13-23. PubMed PMID: 17018289.
34. Chan C, Beltzner CC, Pollard TD. Cofilin dissociates Arp2/3 complex and branches from actin filaments. Curr Biol. 2009;19(7):537-45. PubMed PMID: 19362000.
35. Cairns BR, Erdjument-Bromage H, Tempst P, Winston F, Kornberg RD. Two actin- related proteins are shared functional components of the chromatin-remodeling complexes RSC and SWESNF. Mol Cell. 1998;2(5):639-51. PubMed PMID: 9844636.
36. Zhao K, Wang W, Rando OJ, Xue Y, Swiderek K, Kuo A, Crabtree GR. Rapid and phosphoinositol-dependent binding of the SWESNF-like BAF complex to chromatin after T lymphocyte receptor signaling. Cell. 1998;95(5):625-36. PubMed PMID: 9845365.
37. Hofmann WA, Stojiljkovic L, Fuchsova B, Vargas GM, Mavrommatis E,
Philimonenko V, Kysela K, Goodrich JA, Lessard JL, Hope TJ, Hozak P, de Lanerolle P. Actin is part of pre-initiation complexes and is necessary for transcription by RNA polymerase II. Nat Cell Biol. 2004;6(11): 1094-10T PubMed PMID: 15502823.
38. Han L, Stope MB, de Jesus ML, Oude Weernink PA, Urban M, Wieland T, Rosskopf D, Mizuno K, Jakobs KH, Schmidt M. Direct stimulation of receptor-controlled
phospholipase D1 by phospho-cofilin. Embo J. 2007;26(19):4189-202. PubMed PMID: 17853892.
39. Eibert SM, Lee KH, Pipkorn R, Sester U, Wabnitz GH, Giese T, Meuer SC, Samstag Y. Cofilin peptide homologs interfere with immunological synapse formation and T cell activation. Proc Natl Acad Sci U S A. 2004; 101(7): 1957-62. PubMed PMID: 1476217T 40. Samstag Y, Eibert SM, Klemke M, Wabnitz GH. Actin cytoskeletal dynamics in T lymphocyte activation and migration. J Leukoc Biol. 2003;73(l):30-48. PubMed PM ID
12525560.
41. Samstag Y, Nebl G. Ras initiates phosphatidyl-inositol-3-kinase (PI3K)/PKB mediated signalling pathways in untransformed human peripheral blood T lymphocytes. Adv Enzyme Regul. 2005;45:52-62. PubMed PMID: 16083947.
42. Vorster PJ, Guo J, Yoder A, Wang W, Zheng Y, Xu X, Yu D, Spear M, Wu Y. LIM kinase 1 modulates cortical actin and CXCR4 cycling and is activated by HIV-1 to initiate viral infection. J Biol Chem. 2011;286(14): 12554-64. PubMed PMID: 21321123.
43. Saleh S, Solomon A, Wightman F, Xhilaga M, Cameron PU, Lewin SR. CCR7 ligands CCL19 and CCL21 increase permissiveness of resting memory CD4+ T cells to HIV- 1 infection: a novel model of HIV-1 latency. Blood. 2007;110(13):4161-4. PubMed PMID: 17881634.
44. Guo J, Wang W, Yu D, Wu Y. Spinoculation triggers dynamic actin and cofilin activity facilitating HIV-1 infection of transformed and resting CD4 T cells. J Virol.
2011;85(19):9824-33. PubMed PMID: 21795326.
45. Jimenez-Baranda S, Gomez -Mouton C, Rojas A, Martinez -Prats L, Mira E, Ana Lacalle R, Valencia A, Dimitrov DS, Viola A, Delgado R, Martinez AC, Manes S. Filamin-A regulates actin-dependent clustering of HIV receptors. Nat Cell Biol. 2007;9(7):838-46. PubMed PMID: 17572668.
46. Trushin SA, Bren GD, Badley AD. CXCR4 Tropic HIV-1 gpl20 Inhibition of SDF- 1 alpha-induced Chemotaxis Requires Lck and is Associated with Cofilin Phosphorylation. Open Virol J. 2010;4: 157-62. PubMed PMID: 20835359.
47. Stolp B, Reichman-Fried M, Abraham L, Pan X, Giese SI, Hannemann S, Goulimari P, Raz E, Grosse R, Fackler OT. HIV-1 Nef interferes with host cell motility by deregulation of Cofilin. Cell Host Microbe. 2009;6(2): 174-86. PubMed PMID: 19683683.
48. Nobile C, Rudnicka D, Hasan M, Aulner N, Porrot F, Machu C, Renaud O, Prevost MC, Hivroz C, Schwartz O, Sol-Foulon N. HIV-1 Nef inhibits ruffles, induces filopodia, and modulates migration of infected lymphocytes. J Virol. 2010;84(5):2282-93. PubMed PMID: 20015995. 49. He S, Fu Y, Guo J, Spear M, Yang J, Trinite B, Qin C, Fu S, Jiang Y, Zhang Z, Xu J, Ding H, Levy DN, Chen W, Petricoin E, 3rd, Liotta LA, Shang H, Wu Y. Cofilin
hyperactivation in HIV infection and targeting the cofilin pathway using an anti-alpha4beta7 integrin antibody. Sci Adv. 2019;5(l):eaaf7911. doi: 10.1126/sciadv.aaf7911. PubMed PMID: 30662943; PMCID: PMC6326757.
50. Wu Y, Yoder A, Yu D, Wang W, Liu J, Barrett T, Wheeler D, Schlauch K. Cofilin activation in peripheral CD4 T cells of HIV-1 infected patients: a pilot study. Retrovirology. 2008;5:95. PubMed PMID: 18928553.
51. Santosuosso M, Righi E, Lindstrom V, Leblanc PR, Poznansky MC. HIV-1 envelope protein gpl20 is present at high concentrations in secondary lymphoid organs of individuals with chronic HIV- 1 infection. J Infect Dis. 2009;200(7): 1050-3. PubMed PMID: 19698075.
52. Ramratnam B, Mittler JE, Zhang L, Boden D, Hurley A, Fang F, Macken CA, Perelson AS, Markowitz M, Ho DD. The decay of the latent reservoir of replication- competent HIV-1 is inversely correlated with the extent of residual viral replication during prolonged anti-retroviral therapy. Nat Med. 2000;6(l):82-5. PubMed PMID: 10613829.
53. Pierobon M, Belluco C, Liotta LA, Petricoin EF, 3rd. Reverse phase protein microarrays for clinical applications. Methods Mol Biol. 2011;785:3-12. PubMed PMID: 21901589.
54. Lappalainen P, Drubin DG. Cofilin promotes rapid actin filament turnover in vivo. Nature. 1997;388(6637):78-82. PubMed PMID: 9214506.
55. Yi F, Guo J, Dabbagh D, Spear M, He S, Kehn-Hall K, Fontenot J, Yin Y, Bibian M, Park CM, Zheng K, Park H, Soloveva V, Gharaibeh D, Retterer C, Zamani R, Pitt ML, Naughton J, Jiang Y, Shang H, Hakami RM, Ling B, Young JA, Bavari S, Xu X, Feng Y, Wu Y. Discovery of Novel Small Molecule Inhibitors of LIM Domain Kinase for Inhibiting HIV- 1. J Virol. 2017. doi: 10.1128/JVI.02418-16. PubMed PMID: 28381571.
56. Nishita M, Aizawa H, Mizuno K. Stromal cell-derived factor 1 alpha activates LIM kinase 1 and induces cofilin phosphorylation for T-cell chemotaxis. Molecular & Cellular Biology. 2002;22(3):774-83.
57. Wu Y, Marsh JW. Selective transcription and modulation of resting T cell activity by preintegrated HIV DNA. Science. 2001;293(5534): 1503-6. 58. Wulfkuhle JD, Berg D, Wolff C, Langer R, Tran K, Illi J, Espina V, Pierobon M, Deng J, DeMichele A, Walch A, Bronger H, Becker I, Waldhor C, Etofler H, Esserman L, Investigators 1ST, Liotta LA, Becker KF, Petricoin EF, 3rd. Molecular analysis of HER2 signaling in human breast cancer by functional protein pathway activation mapping. Clin Cancer Res. 2012;18(23):6426-35. doi: 10.1158/1078-0432.CCR-12-0452. PubMed PMID:
23045247.

Claims

What is claimed is
1. A method for detecting and treating immune dysfunction in a patient, comprising
(a) quantifying cofilin phosphorylation levels in T cells (CD4/CD8),
monocytes/macrophages, B cells, natural killer (NK) cells, and/or cancer cells of said patient, wherein said patient with immune dysfunction has lower or higher cofilin phosphorylation levels than a control healthy person or patient; and
(b) administering to said patient an effective amount of a composition that corrects cofilin dysregulation and restores cofilin phosphorylation.
2. The method of claim 1, wherein said patient has a chronic disease causing immune dysfunction.
3. The method of claim 2, wherein said chronic disease is HIV infection or cancer.
4. The method of claim 1, wherein said immune dysfunction is aberrant T cell,
macrophage, B cell, NK cell activation, impairment of T cell, monocyte/macrophage, B cell, NK cell motility and/or tissue migration.
5. The method of claim 1, wherein said composition is a cofilin phosphatase inhibitor or cofilin kinase activator.
6. The method of claim 5, wherein said composition comprises okadaic acid.
7. A method for determining immune dysfunction in a patient, comprising quantifying cofilin phosphorylation in T cells (CD4/CD8), monocyte/macrophages, B cells, natural killer cells, and/or cancer cells in said patient.
8. The method of claim 7, wherein said patient is a cancer patient or HIV-infected
patient.
9. A method for treating cancer in a patient, comprising
(a) quantifying cofilin phosphorylation levels in T cells (CD4/CD8), monocyte/macrophages, B cells, natural killer cells, and/or cancer cells of said patient, wherein said patient has lower or higher cofilin phosphorylation levels than a control healthy person; and
(b) administering to said patient an effective amount of a composition that restores cofilin phosphorylation.
10. The method of claim 9, wherein said composition is a cofilin phosphatase inhibitor or cofilin kinase activator or an antibody against chemotactic receptors that can trigger cofilin phosphorylation or dephosphorylation or a small molecule that can trigger cofilin phosphorylation or dephosphorylation.
11. The method of claim 10, wherein said composition comprises okadaic acid.
12. Any method for treatment of a patient with cancer disclosed herein.
13. The method of claim 12, wherein the patient does not have AIDS.
14. The method of claim 12, wherein the patient does not have HIV.
PCT/US2020/014542 2019-01-23 2020-01-22 Cofilin phosphorylation for cancer treatment WO2020154359A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US18/118,409 US20230248907A1 (en) 2019-07-11 2023-03-07 Infusion Unit

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201962795873P 2019-01-23 2019-01-23
US62/795,873 2019-01-23

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US18/118,409 Continuation-In-Part US20230248907A1 (en) 2019-07-11 2023-03-07 Infusion Unit

Publications (1)

Publication Number Publication Date
WO2020154359A1 true WO2020154359A1 (en) 2020-07-30

Family

ID=71736987

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2020/014542 WO2020154359A1 (en) 2019-01-23 2020-01-22 Cofilin phosphorylation for cancer treatment

Country Status (1)

Country Link
WO (1) WO2020154359A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009155310A1 (en) * 2008-06-17 2009-12-23 University Of Rochester Tumor suppression through plexin c1
WO2010095042A2 (en) * 2009-02-20 2010-08-26 Centre Natonal De La Recherche Scientifique Pyridocarbazole type compounds and applications thereof
WO2014002101A1 (en) * 2012-06-28 2014-01-03 Ramot At Tel-Aviv University Ltd Lim kinase inhibitors
US8802649B2 (en) * 2004-05-06 2014-08-12 University Of Central Florida Research Foundation, Inc. Methods and compositions for inhibiting the proliferation of cancer cells
WO2019147678A1 (en) * 2018-01-23 2019-08-01 Virongy L.L.C. Cofilin phosphorylation for quantifying cd4 t cell damage and predict cd4 t cell recovery from anti-retroviral therapy

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8802649B2 (en) * 2004-05-06 2014-08-12 University Of Central Florida Research Foundation, Inc. Methods and compositions for inhibiting the proliferation of cancer cells
WO2009155310A1 (en) * 2008-06-17 2009-12-23 University Of Rochester Tumor suppression through plexin c1
WO2010095042A2 (en) * 2009-02-20 2010-08-26 Centre Natonal De La Recherche Scientifique Pyridocarbazole type compounds and applications thereof
WO2014002101A1 (en) * 2012-06-28 2014-01-03 Ramot At Tel-Aviv University Ltd Lim kinase inhibitors
WO2019147678A1 (en) * 2018-01-23 2019-08-01 Virongy L.L.C. Cofilin phosphorylation for quantifying cd4 t cell damage and predict cd4 t cell recovery from anti-retroviral therapy

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
AMBACH, A ET AL.: "The serine phosphatases PP1 and PP2A associate with and activate the actin-binding protein cofiiin in human T iymphocytes", EUROPEAN JOURNAL OF IMMUNOLGY, vol. 30, no. 12, December 2000 (2000-12-01), pages 3422 - 3431, XP002222451 *
HE, S ET AL.: "C ofilin hyperactivation in HIV infection and targeting the cofilin pathway using an anti-alpha4-alpha7 integrin antibody", SCIENCE ADVANCES, vol. 5, no. 1, 9 January 2019 (2019-01-09), pages 1 - 12, XP55727832 *
PRUDENT, R ET AL.: "A quantitative proteomic analysis of cofilin phosphorylation in myeloid cells and its modulation using the LIM kinase inhibitor Pyr1", PL .OS ONE, vol. 13, no. 12, 14 December 2018 (2018-12-14), pages 1 - 17, XP55727833 *

Similar Documents

Publication Publication Date Title
Lammers et al. Identification of a novel immunomodulatory gliadin peptide that causes interleukin‐8 release in a chemokine receptor CXCR3‐dependent manner only in patients with coeliac disease
Günther et al. Angiotensin receptor type 1 and endothelin receptor type A on immune cells mediate migration and the expression of IL-8 and CCL18 when stimulated by autoantibodies from systemic sclerosis patients
Steiner et al. Deficiency in coatomer complex I causes aberrant activation of STING signalling
Uto-Konomi et al. CXCR7 agonists inhibit the function of CXCL12 by down-regulation of CXCR4
Fosbrink et al. C5b-9-induced endothelial cell proliferation and migration are dependent on Akt inactivation of forkhead transcription factor FOXO1
Cheung et al. An arrestin-dependent multi-kinase signaling complex mediates MIP-1β/CCL4 signaling and chemotaxis of primary human macrophages
Wilson et al. Mini ReviewThe Effect of Interleukin-16 and its Precursor on T Lymphocyte Activation and Growth
Messmer-Blust et al. The Interferon-γ–induced Murine Guanylate-Binding Protein-2 Inhibits Rac Activation during Cell Spreading on Fibronectin and after Platelet-derived Growth Factor Treatment: Role for Phosphatidylinositol 3-Kinase
Tocheva et al. Quantitative phosphoproteomic analysis reveals involvement of PD-1 in multiple T cell functions
Campbell et al. CCL2 increases X4-tropic HIV-1 entry into resting CD4+ T cells
Zhou et al. GDF6-CD99 signaling regulates src and ewing sarcoma growth
Huang et al. HMGB1 induces endothelial progenitor cells apoptosis via RAGE-dependent PERK/eIF2α pathway
Boeck et al. The human cytomegalovirus US27 gene product constitutively activates antioxidant response element-mediated transcription through Gβγ, phosphoinositide 3-kinase, and nuclear respiratory factor 1
Bae et al. Trp-Lys-Tyr-Met-Val-D-Met is a chemoattractant for human phagocytic cells
Jayabal et al. NELL2-cdc42 signaling regulates BAF complexes and Ewing sarcoma cell growth
US7691582B2 (en) Methods of secretory vimentin detection and modulation
Dasgupta et al. Mechanism of activation-induced downregulation of mitofusin 2 in human peripheral blood T cells
US20210041437A1 (en) Cofilin phosphorylation for quantifying cd4 t cell damage and predict cd4 t cell recovery from anti-retroviral therapy
Ding et al. Silica-exposed macrophages-secreted exosomal miR125a-5p induces Th1/Th2 and Treg/Th17 cell imbalance and promotes fibroblast transdifferentiation
Wiiger et al. The epidermal growth factor receptor (EGFR) and proline rich tyrosine kinase 2 (PYK2) are involved in tissue factor dependent factor VIIa signalling in HaCaT cells
Foley et al. Differentiation of human T cells alters their repertoire of G protein α-subunits
WO2020154359A1 (en) Cofilin phosphorylation for cancer treatment
Mangan et al. Clostridium difficile Toxin B activates the NLRP3 inflammasome in human macrophages, demonstrating a novel regulatory mechanism for the Pyrin inflammasome
KR101640326B1 (en) Map4k3 as a biomarker and therapeutic target for autoimmune disease, cancer, inflammation and il-17-associated disease
He et al. Cofilin hyperactivation in HIV infection and targeting the cofilin pathway using an anti- 4 7 integrin antibody

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 20745582

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 20745582

Country of ref document: EP

Kind code of ref document: A1