R M Rerko's research while affiliated with Case Western Reserve University School of Medicine and other places

What is this page?


This page lists the scientific contributions of an author, who either does not have a ResearchGate profile, or has not yet added these contributions to their profile.

It was automatically created by ResearchGate to create a record of this author's body of work. We create such pages to advance our goal of creating and maintaining the most comprehensive scientific repository possible. In doing so, we process publicly available (personal) data relating to the author as a member of the scientific community.

If you're a ResearchGate member, you can follow this page to keep up with this author's work.

If you are this author, and you don't want us to display this page anymore, please let us know.

Publications (12)


Treatment with Th1-deviating rIL-12 and anti–IL-4 antibodies or transient depletion of CD4⁺ T cells fails to cure established murine leishmaniasis in susceptible BALB/c mice. (A) Combined treatment with neutralizing anti–IL-4 mAb (11B11) and rIL-12 does not reverse established infection. BALB/c mice (n = 6 per group) were infected with L. major and treated with (Control) 0.5 mg of rat IgG administered on days 7, 14, and 21 of infection (control) or 1.0 μm of rIL-12 (Hoffmann-LaRoche; 2.1 × 10³ U/dose) injected intraperitoneally daily for 14 d starting on day 7 of infection. A separate group of mice (11B11/rIL-12) were injected intraperitoneally on days 7, 14, and 21 with 1.0 mg of neutralizing anti–IL-4 mAb 11B11 in combination with rIL-12 for 14 d. Shown are mean footpad thicknesses ± SEM. Although footpad swelling was significantly suppressed, all mice eventually progressed to develop ulceration and deep tissue necrosis. (B) Separate groups of BALB/c mice (n = 5 each) were infected with L. major for 3 wk and treated on days 21 and 22 with rat IgG (control) or with 0.33 mg of anti-CD4 mAb GK1.5 by intraperitoneal injection. Another group (11B11/rIL-12) was treated on day 21 with 1.0 mg of anti–IL-4 mAb 11B11 and then with 0.5 μg of rIL-12 given by intralesional injection into each footpad (Hoffmann-LaRoche; 1.1 × 10³ U/dose) for 8 consecutive days. All mice progressed to develop ulceration and necrosis.
BALB/c mice infected for 3 wk with L. major can be cured by CD4⁺ depletion and Th1-deviating cytokine therapy. Groups of five BALB/c mice were treated with (GK1.5/11B11/rIL-12) 0.5 mg of GK1.5 mAb and 11B11 anti–IL-4 mAb given daily for 2 d, followed by a total of 10 intralesional injections of 0.5 μg of rIL-12 (Hoffmann-LaRoche; 1.1 × 10³ U/dose) into each foot from days 21 through 32 or 0.5 mg of rat IgG and intralesional saline only (control). Data represent mean footpad thickness ± SEM measured weekly by micrometer. All control mice were killed at 8 wk because of progressive cutaneous ulceration and necrosis; five out of five mice receiving GK1.5 immunotherapy healed. At 14 wk of infection, the five BALB/c mice cured by immunotherapy and five normal BALB/c mice (reinfection control) were injected with 2 × 106 L. major promastigotes per footpad and footpad thicknesses recorded weekly.
Specific depletion of CD4⁺ T cells is required for cure of established leishmaniasis in response to treatment with rIL-12 and anti–IL-4 mAb. Groups of five BALB/c mice were infected with L. major for 3 wk and then injected on 2 consecutive d with 0.5 mg of rat IgG (control), depleting anti-CD4 mAb GK1.5 (GK1.5), depleting anti-CD8 mAb (Lyt2.2), or nondepleting anti-CD4 mAb YTS177.9 (YTS). All groups except for controls were subsequently treated intraperitoneally with 1.0 mg of anti–IL-4 mAb 11B11 on day 21 and 1.0 μg of rIL-12 (1.1 × 10³ U/dose) injected into each footpad daily for 8 d. Data represent footpad thicknesses (in millimeters) measured weekly and displayed as mean ± SEM. With the exception of GK1.5-treated mice, all groups developed progressive ulceration and footpad necrosis.
Early and delayed effects of GK1.5-based immunotherapy on lymph node cytokine responses. Groups of six BALB/c mice each were infected with L. major for 3 wk and treated with rat IgG alone (control) or GK1.5 and anti–IL-4 mAbs combined with intralesional rIL-12 as described in Fig. 2 (GK1.5/11B11/rIL-12). Lymph node cells were harvested at 7 and 21 d after the start of therapy and cultured in DMEM/10% FBS alone or in the presence of 10 μg/ml of soluble leishmania antigen. Anti– IL-4 receptor mAb (M-1) was added at 10 μg/ml in duplicate cultures to measure total production of IL-4. After 48 h of culture, supernatants were assayed by ELISA. Data represent mean cytokine concentrations (ng/ml) ± SEM for IL-4 (top) and IFN-γ (bottom). IL-4 production was decreased significantly (P < 0.05) for immunotherapy mice compared with control mice for all times and conditions. IFN-γ production increased significantly (P < 0.05) for immunotherapy mice only during wk 1.
CD4⁺ depletion and cytokine therapy restores Th1 type mRNA responses in vivo while suppressing IL-4 mRNA expression. RNA was harvested from lymph node tissues taken from control (C) and GK1.5/11B11/rIL-12-treated (ITx) mice at 7 and 21 d after the start of immunotherapy. Reverse transcriptase PCR was used to compare expression of selected immune response genes. Mice had been infected previously for 3 wk with L. major. Data represent the expression of the indicated mRNA species as determined by densitometric analysis of gel products obtained by semiquantitative reverse transcriptase PCR. Data are normalized for expression of HPRT in the same sample. Expression of IL-4, IFN-γ, IL-12 receptor β2, and iNOS mRNA was not detectable in naive lymph node tissue at the level of amplification used in these analyses. Primers used for amplification of IL-12 receptor β2 cDNA were (5′ to 3′): GCACAGACTGTTAGAGAATGC (sense) and CCTTCCTGGACACATGATATG (antisense). Primers for murine iNOS cDNA amplification were: TCACGCTTGGGTCTTGTTCACT (sense) and TTGTCTCTGGGTCCTCTGGTCA (antisense).

+1

Cure of Progressive Murine Leishmaniasis: Interleukin 4 Dominance Is Abolished by Transient CD4 T Cell Depletion and T Helper Cell Type 1–selective Cytokine Therapy
  • Article
  • Full-text available

June 1999

·

25 Reads

·

29 Citations

Journal of Experimental Medicine (JEM)

Journal of Experimental Medicine (JEM)

Frederick P. Heinzel

·

Ronald M. Rerko

Progressive infection with Leishmania major in susceptible BALB/c mice is mediated by interleukin (IL)-4-producing T helper cell type 2 (Th2) CD4(+) T cells that, once established, become resistant to Th1-deviating therapies with recombinant (r)IL-12 and/or neutralizing anti-IL-4 antibodies. We sought to restore protective immunity in advanced leishmaniasis by depletion of Th2-biased CD4(+) populations and by cytokine-directed reconstitution of Th1 cellular responses during lymphocyte recovery. Treatment with cytolytic GK1.5 anti-CD4 mAb alone did not reverse disease in 3 wk-infected BALB/c mice, but GK1.5 combined with anti-IL-4 antibody and intralesional rIL-12 cured cutaneous lesions in 80% of mice and established a Th1-polarized cytokine response to L. major antigen protective against reinfection. The curative effects of GK1.5 were not replaced by cytotoxic anti-CD8 monoclonal antibody 2.43 or nondepleting anti-CD4 mAb YTS177, confirming that depletion of CD4(+) cells was specific and essential for therapeutic effect. Finally, combined CD4(+) depletion and IL-4 neutralization were curative, indicating that neither increased parasite burden nor altered accessory cell function independently biased towards Th2 reconstitution in advanced leishmaniasis. Advanced leishmaniasis can be cured by T cell depletion and cytokine-directed recovery of Th1 cellular responses, suggesting novel interventions for other immune-mediated diseases and identifying distinct roles for CD4(+) T cell and non-T cell in the maintenance of Th2 and Th1 phenotypes.

Download
Share

Increased Capacity for Interleukin-2 Synthesis Parallels Disease Progression in Mice Infected with Leishmania major

October 1998

·

12 Reads

·

7 Citations

Lymph node cells of BALB/c mice with progressive leishmaniasis produced sixfold more interleukin-2 (IL-2) in culture than those of healing C57BL/6 mice. IL-2 synthesis also increased in C57BL/6 mice made susceptible by IL-12 or gamma interferon deficiency. However, IL-2 mRNA levels in vivo did not reflect IL-2 production in vitro. Because IL-2 contributes to the pathogenesis of progressive leishmaniasis, the functional significance of these findings should be further explored.


Increased Capacity for Interleukin-2 Synthesis Parallels Disease Progression in Mice Infected withLeishmania major

September 1998

·

2 Reads

·

4 Citations

Infection and Immunity

Infection and Immunity

Lymph node cells of BALB/c mice with progressive leishmaniasis produced sixfold more interleukin-2 (IL-2) in culture than those of healing C57BL/6 mice. IL-2 synthesis also increased in C57BL/6 mice made susceptible by IL-12 or gamma interferon deficiency. However, IL-2 mRNA levels in vivo did not reflect IL-2 production in vitro. Because IL-2 contributes to the pathogenesis of progressive leishmaniasis, the functional significance of these findings should be further explored.


Underproduction of Interleukin-12 in Susceptible Mice during Progressive Leishmaniasis Is Due to Decreased CD40 Activity

April 1998

·

8 Reads

·

52 Citations

Cellular Immunology

Interleukin-12 promotes Th1 lymphocyte responses necessary for the cure of murine Leishmania major infection. We found that IL-12 p40 mRNA expression peaked at 4 weeks of infection in resistant C57BL/6 mice at levels threefold greater than in BALB/c mice. Peak IL-12 p40 expression in both strains was reduced threefold following treatment with neutralizing anti-CD40 ligand antibody and disease worsened in C57BL/6 mice. Direct activation of cultured lymph node cells by anti-CD40 MAb or soluble CD40 ligand failed to restore deficient IL-12 production by infected BALB/c mice unless recombinant IFN-beta was added to culture. Infected BALB/c lymph nodes also contained two- to threefold fewer low-density CD40+ accessory cells compared to that in C57BL/6 mice. We conclude that CD40-dependent responses are continually required for healing of leishmaniasis and that progressive disease is associated with decreased CD40-stimulated IL-12 synthesis as a consequence of either altered cytokine environment or inadequate accessory cell number.


In Vivo Production and Function of IL-12 p40 Homodimers

May 1997

·

22 Reads

·

241 Citations

The Journal of Immunology

The bioactivity of IL-12 is mediated by heterodimers of disulfide-linked p35 and p40 protein subunits. Homodimeric p40 competes with heterodimer for binding to the high affinity IL-12R and inhibits IL-12 bioactivity in vitro. However, the production and significance of p40 homodimer as a cytokine antagonist in vivo have not been determined. In these studies, we observed increased amounts of both IL-12 p40 monomer and homodimer in the serum of C57BL/6 mice following injection of 300 microg of Salmonella enteritidis LPS. Homodimer constituted between 20 and 40% of the total circulating p40 in endotoxemic sera, as confirmed by both Sephacryl S-100 gel filtration and p40-specific immunoprecipitation analyses. Similar relative amounts of homodimer and monomer were observed in endotoxemic BALB/c, C57BL/6, IFN-gamma-deficient C57BL/6 mice and C57BL/6 mice previously infected with bacille Calmette-Guérin. To determine whether IL-12 p40 homodimer was capable of antagonizing IL-12-dependent IFN-gamma responses in vivo, we pretreated C57BL/6 mice with purified rIL-12 p40 homodimer before i.p. challenge with endotoxin. Mice treated with 40 to 80 microg of p40 homodimer generated 80 to 82% less circulating IFN-gamma during acute endotoxemia than saline controls (p < 0.01). We conclude that p40 homodimer is produced in vivo, functions as a cytokine antagonist in the context of the mouse model of acute endotoxemia, and may represent a novel form of self-regulating cytokine response.


IFN-γ-Independent Production of IL-12 during Murine Endotoxemia

December 1996

·

9 Reads

·

52 Citations

The Journal of Immunology

We examined whether IFN-gamma deficiency alters the in vivo IL-12 response occurring in the mouse model of acute endotoxemia. C57BL/6 IFN-gamma knockout mice (IFN-gamma 0/0) produced as much circulating IL-12 p40 and IL-12 p70 as did IFN-gamma +/+ mice following injection with S. enteritidis LPS, despite sustaining 11-fold reductions in circulating TNF-alpha. Pretreatment of IFN-gamma 0/0 mice with recombinant mouse IFN-gamma (rIFN-gamma) enhanced circulating TNF-alpha by as much as sixfold, but serum IL-12 p40 and IL-12 p70 responses increased by only twofold or less. Compared with IFN-gamma +/+ mice, the spleens of endotoxemic IFN-gamma 0/0 mice generated two- to threefold fewer IL-12 p40-secreting cells following in vitro or in vivo exposure to endotoxin. The addition of rIFN-gamma to IFN-gamma 0/0 splenocyte culture restored normal levels of LPS-stimulated IL-12 p40 production. Removal of Mac-1+ or F4/80+ cells from endotoxin-stimulated spleen reduced both TNF-alpha and IL-12 p40 production, but 33D1+ dendritic cell removal only affected IL-12 synthesis. These data suggest that the aggregate IL-12 p40 and p70 response to endotoxemia in vivo is IFN-gamma-independent and distinct from IFN-gamma-dependent serum TNF-alpha and splenic IL-12 responses. The cellular and/or cytokine basis for the unexpected preservation of IL-12 production in IFN-gamma-deficient mice may be relevant to normal and pathologic immune responses.


Immunoregulation of murine leishmaniasis by interleukin-12

September 1995

·

7 Reads

·

16 Citations

Research in Immunology

Distinct phenotypic outcomes following infection of mice with Leishmania major are closely linked to the emergence of functionally dissimilar Th1 or Th2 CD4+ T-cell responses early in the course of disease. This model of T-cell-dependent microbial pathology has proven useful for the study of cytokine regulatory and effector functions in vivo. To this end, the causal relationships linking synthesis of IFN gamma to cure and of IL4 to disease exacerbation have already been well characterized. IL12 also has a defined role in shaping the immune response against L. major. Early treatment with recombinant IL12, or vaccination using IL12 as an adjuvant, protects genetically susceptible hosts from progressive infection. Protective mechanisms include both suppression of deleterious Th2 cell responses and amplification of beneficial Th1 cell activities. Although Leishmania are poor stimuli for macrophage-derived IL12 when compared to bacteria and other protozoa, in vivo production during infection can be indirectly demonstrated by the worsening of leishmaniasis that follows anti-IL12 injection in normally resistant mice. Whether IL12 production during infection represents constitutive or regulated synthesis by infected macrophages is unresolved and deserves further exploration.


Endogenous IL-12 is required for control of Th2 cytokine responses capable of exacerbating leishmaniasis in normally resistant mice

August 1995

·

8 Reads

·

172 Citations

The Journal of Immunology

We investigated the mechanisms by which treatment with anti-IL-12 Ab prevents cure of infection with Leishmania major in resistant C57BL/6 mice. Consistent with delayed production of IL-12, anti-IL-12 Abs could be administered as late as 2 wk after infection to exacerbate disease. Starting at 2 wk of infection, the cultured lymph node cells from mice treated with either polyclonal or monoclonal anti-IL-12 Abs persistently generated 3- to 10-fold more IL-4 and IL-10 in response to L. major Ag compared with cells from mice receiving preimmune goat IgG. Reciprocal decreases in Ag-specific IFN-gamma production were observed in mice receiving anti-IL-12 Abs. A similar reversal of IFN-gamma and IL-4 production accompanied progressive disease induced by pretreatment with a single dose of anti-IFN-gamma mAb. Although IFN-gamma production was suppressed for up to 4 wk in mice treated with monoclonal anti-IL-12 or anti-IFN-gamma, coadministration of neutralizing anti-IL-4 IgG reversed progressive illness. These findings demonstrate that IL-12 produced in vivo is necessary for both the emergence of IFN-gamma producing cells and the down-regulation of Th2 cell responses during murine leishmaniasis. Furthermore, the uninhibited production of IL-4 was required to sustain progressive infection initiated by the decreased IFN-gamma synthesis observed in anti-IL-12 and anti-IFN-gamma-treated mice.


Interleukin 12 is produced in vivo during endotoxemia and stimulates synthesis of gamma interferon

November 1994

·

70 Reads

·

238 Citations

Infection and Immunity

Infection and Immunity

F P Heinzel

·

R M Rerko

·

·

[...]

·

D S Schoenhaut

Gamma interferon (IFN-gamma) is produced in response to circulating lipopolysaccharide (LPS) and contributes to the lethality of endotoxic shock. To address the cellular source of IFN-gamma production in vivo, T cells and B cells were magnetically purified from C57BL/6 mouse spleens 5 h following endotoxin injection. IFN-gamma RNA was abundant in splenic CD4+ and CD8+ T cells and in a T- and B-cell-depleted population of splenocytes containing 34% NK1.1+ natural killer (NK) cells. Because interleukin 12 (IL-12) is a known inducer of IFN-gamma synthesis by cultured T cells and NK cells, we examined whether IL-12 might be involved in IFN-gamma release during endotoxemia. mRNA encoding the p40 subunit of IL-12 increased markedly in the spleens of C57BL/6 mice at 2 h after LPS injection, whereas p35 IL-12 mRNA was constitutively expressed at all times. Bioactive IL-12 (p70 heterodimer) was detected in mouse serum at 2 to 4 h after LPS injection. Similar results were obtained using a p40 subunit-specific enzyme-linked immunosorbent assay. Endotoxin-insensitive C3H/HeJ mice generated threefold less IL-12 p70 and IFN-gamma at these times than endotoxin-sensitive C3H/HeOuJ mice. Pretreatment of mice with polyclonal anti-mouse IL-12 antibody reduced IFN-gamma levels present at 6 h post-LPS nearly sixfold in three separate experiments. These studies support a role for IL-12 as a proximal stimulator of IFN-gamma release during endotoxemia.


Recombinant interleukin 12 cures mice infected with Leishmania major

June 1993

·

37 Reads

·

676 Citations

Journal of Experimental Medicine (JEM)

Journal of Experimental Medicine (JEM)

Resistant C57BL/6 mice infected with Leishmania major are self-healing, whereas susceptible BALB/c mice fail to contain cutaneous infection and subsequently undergo fatal visceral dissemination. These disparate outcomes are mediated by dissimilar expansions of T helper type 1 (Th1) and Th2 CD4+ T lymphocyte subsets in vivo during cure and progression of disease. Because interleukin 12 (IL-12) has potent T cell growth and interferon gamma (IFN-gamma) stimulatory effects, we studied its effect on CD4+ T cell differentiation during murine leishmaniasis. Treatment with recombinant murine (rMu)IL-12 during the first week of infection cured 89% of normally susceptible BALB/c mice, as defined by decreased size of infected footpads and 1,000-10,000-fold reduced parasite burdens, and provided durable resistance against reinfection. Cure was associated with markedly depressed production of IL-4 by lymph node cells cultured with antigen or mitogen, but preserved or increased production of IFN-gamma relative to untreated mice. IL-4 and IFN-gamma mRNA associated with CD4+ T lymphocytes isolated from infected lymph nodes showed similar reciprocal changes in response to rMuIL-12 therapy. A single injection of anti-IFN-gamma monoclonal antibody abrogated the protective effect of rMuIL-12 therapy and restored Th2 cytokine responses. We conclude that rMuIL-12 prevents deleterious Th2 T cell responses and promotes curative Th1 responses in an IFN-gamma-dependent fashion during murine leishmaniasis. Since BALB/c leishmaniasis cannot be cured with rMuIFN-gamma alone, additional direct effects of IL-12 during T cell subset selection are suggested. Because rMuIL-12 is uniquely protective in this well-characterized model of chronic parasitism, differences in IL-12 production may underlie heterogenous host responses to L. major and other intracellular pathogens.


Citations (10)


... There are well documented reports that, in IL-10 (-/-) mice, protective Th1 responses are stimulated by IL-12 derived from activated MФ. This synergism between IFNγ and IL-12 further enhances protective immunity against VL [63,64]. In addition, IL-12 favors Th1 immune response by escalating NOS-2 expression and T cell proliferation [65]. ...

Reference:

Dipeptidylcarboxypeptidase of Leishmania donovani: A potential vaccine molecule against experimental visceral leishmaniasis
Endogenous IL-12 is required for control of Th2 cytokine responses capable of exacerbating leishmaniasis in normally resistant mice
  • Citing Article
  • August 1995

The Journal of Immunology

... IL-12α potentiates IFN-γ-mediated signal via induction of IFN-γ secretion within different cell types [57]. As anticipated, CS + Tx induced IFN-γ and IL-12α expression in renal grafts, both of which regulate IFN-γ expression [101]. IFN-γ mediates its pro-inflammatory signal by activating STAT1 transcription factors and through its additional macrophage-stimulating properties [102,103]. ...

Interleukin 12 is produced in vivo during endotoxemia and stimulates synthesis of gamma interferon
Infection and Immunity

Infection and Immunity

... DCs initiate the adaptive response to Leishmania. IL-12, produced by DCs in response to NK cell-derived IFN γ, is essential for the development of TH1 CD4 + cells that have anti-Leishmania immune resistance [152,153] . TH1 CD4 + cells, recruited to the cutaneous lesions, produce IFNγ that activates macrophages and promotes the killing of Leishmania parasites [103] . ...

Recombinant interleukin 12 cures mice infected with Leishmania major
Journal of Experimental Medicine (JEM)

Journal of Experimental Medicine (JEM)

... Patients with recurrent CL presented the cytokines IL-2, IL-4, and TNF correlated with each other. The relation between IL-2 and TNF has already been elucidated, however, IL-2 also can stimulate the proliferation of Th2 cells through the production of IL-4 and the association between these cytokines had already been demonstrated in BALB/c mice infected with L. major (Heinzel et al., 1993;Maspi et al., 2016). The correlation between TNF and IL-4 was surprising since TNF mediates Th1 response. ...

IL-2 is necessary for the progression of Leishmaniasis in susceptible murine hosts
  • Citing Article
  • June 1993

The Journal of Immunology

... A notable feature of immunization induced in the mice receiving the SP15 family salivary proteins as a booster was documented by a greater percentage of splenocytes producing IL-12 by CD14 + cells and IFN-γ by CD4 + cells. A previous report shows that IL-12 is a potent TH 1-inducing adjuvant [19]. Thus, the increased percentage of CD4 + T cells producing IFN-γ is interpreted as a positive effect in vaccinated hosts. ...

Immunoregulation of murine leishmaniasis by interleukin-12
  • Citing Article
  • September 1995

Research in Immunology

... Discrepancies in Il12a and Ifnγ expression in ASF-colonized mice after SFB colonization suggest an IFNγ-independent production of IL12. Production of IL12 in an IFNγ-independent manner is hypothesized to be relevant for physiological and pathological immune responses (62). However, whether particular bacterial species play a role in downregulation of Ifnγ expression in chronic MNV infection needs to be further investigated. ...

IFN-γ-Independent Production of IL-12 during Murine Endotoxemia
  • Citing Article
  • December 1996

The Journal of Immunology

... The p40 family of cytokines has four members, p40 homodimer (p40 2 ), p40 monomer (p40), IL-12 (p40:p35), and IL-23 (p40:p19) [8]. IL-12 plays a crucial role in the early inflammatory response by producing T helper type 1 (Th1) cells [9,10]. It is believed that most effects are mediated by IFN-γ secretion [11]. ...

In Vivo Production and Function of IL-12 p40 Homodimers
  • Citing Article
  • May 1997

The Journal of Immunology

... L. donovani infection blocks LPS-mediated B7-1 expression in infected macrophages [211,212]. In the case of infection with L. amazonensis, disruption of CD40/CD40L ligation results in enhanced susceptibility [213] through inhibition of iNOS expression [213,214] and IL-12 production [215] by infected macrophages. Macrophages infected with L. major also show defects in CD40 signalling in a p38-dependent manner [216]. ...

Underproduction of Interleukin-12 in Susceptible Mice during Progressive Leishmaniasis Is Due to Decreased CD40 Activity
  • Citing Article
  • April 1998

Cellular Immunology

... The parasite also blocks the physiologic role of IL2. Many studies indicated that BALB/c mice are more sensitive to Leishmania infection than C57/BALB mice due to suppressed IL2 production and IL2-R reduction (Frederick et al., 1998). IL2 cytokine stimulates proliferation and differentiation of T lymphocytes and triggers cell growth cycle of T cells. ...

Increased Capacity for Interleukin-2 Synthesis Parallels Disease Progression in Mice Infected with Leishmania major
  • Citing Article
  • October 1998

Infection and Immunity

Infection and Immunity

... Previous studies suggested that CD4 + t cell depletion skews the immune response toward type 1 polarization [42]. However, in the present study, IFn-γ and the IFn-γ-dependent th1 chemokines CXCl9 and CXCl10 increased in mouse serum during combination It, but not after anti-CD4 mab or rIl-21 treatment as single agents. ...

Cure of Progressive Murine Leishmaniasis: Interleukin 4 Dominance Is Abolished by Transient CD4 T Cell Depletion and T Helper Cell Type 1–selective Cytokine Therapy
Journal of Experimental Medicine (JEM)

Journal of Experimental Medicine (JEM)